CA2924141C - 5-amino 4-cyano substituted oxazole and thiazole derivatives as inhibitors of human 12/15-lipoxygenase - Google Patents

5-amino 4-cyano substituted oxazole and thiazole derivatives as inhibitors of human 12/15-lipoxygenase Download PDF

Info

Publication number
CA2924141C
CA2924141C CA2924141A CA2924141A CA2924141C CA 2924141 C CA2924141 C CA 2924141C CA 2924141 A CA2924141 A CA 2924141A CA 2924141 A CA2924141 A CA 2924141A CA 2924141 C CA2924141 C CA 2924141C
Authority
CA
Canada
Prior art keywords
lox
carbonitrile
lipoxygenase
compound
oxazole
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
CA2924141A
Other languages
French (fr)
Other versions
CA2924141A1 (en
Inventor
Klaus Van Leyen
Theodore R. Holman
David J. Maloney
Ajit Jadhav
Anton Simeonov
Ganesha RAI
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
US Department of Health and Human Services
General Hospital Corp
University of California Santa Cruz
Original Assignee
US Department of Health and Human Services
General Hospital Corp
University of California Santa Cruz
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by US Department of Health and Human Services, General Hospital Corp, University of California Santa Cruz filed Critical US Department of Health and Human Services
Publication of CA2924141A1 publication Critical patent/CA2924141A1/en
Application granted granted Critical
Publication of CA2924141C publication Critical patent/CA2924141C/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/04Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/42Oxazoles
    • A61K31/4211,3-Oxazoles, e.g. pemoline, trimethadione
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/42Oxazoles
    • A61K31/422Oxazoles not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/4245Oxadiazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/425Thiazoles
    • A61K31/4261,3-Thiazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4709Non-condensed quinolines and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/472Non-condensed isoquinolines, e.g. papaverine
    • A61K31/4725Non-condensed isoquinolines, e.g. papaverine containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D213/78Carbon atoms having three bonds to hetero atoms, with at the most one bond to halogen, e.g. ester or nitrile radicals
    • C07D213/84Nitriles
    • C07D213/85Nitriles in position 3
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/28Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to ring carbon atoms
    • C07D239/32One oxygen, sulfur or nitrogen atom
    • C07D239/42One nitrogen atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D263/00Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings
    • C07D263/02Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings not condensed with other rings
    • C07D263/30Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D263/34Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D263/48Nitrogen atoms not forming part of a nitro radical
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D277/00Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings
    • C07D277/02Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings
    • C07D277/20Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D277/32Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D277/38Nitrogen atoms
    • C07D277/42Amino or imino radicals substituted by hydrocarbon or substituted hydrocarbon radicals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D277/00Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings
    • C07D277/02Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings
    • C07D277/20Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D277/32Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D277/56Carbon atoms having three bonds to hetero atoms with at the most one bond to halogen

Abstract

A systematic screening has revealed a family of compounds that exhibit inhibitory effects on 12/15-lipoxygenase. Accordingly, the present invention relates to the use of these compounds for the inhibition of 12/15-lipoxygenase and for the treatment of a condition involving 12/15-lipoxygenase. Exemplary conditions include, but are not limited to, stroke, periventricular leukomalacia, cardiac arrest with resuscitation, atherosclerosis, Parkinson's disease, Alzheimer's disease, and breast cancer.

Description

5-amino 4-cyano substituted oxazole and thiazole derivatives as inhibitors of human 12/15-lipoxygenase Pon [00021 TECHNICAL FIELD
[00031 The present invention relates generally to inhibition of 12/15-lipoxygenase, treatment of a condition involving 12/15-lipoxygenase including, but not limited to, strokes, periventricular leukomalacia, cardiac arrest with resuscitation, atherosclerosis, Parkinson's disease, Alzheimer's disease, and breast cancer.
BACKGROUND
[00041 Classical drug discovery to a large extent relies on animal models of disease, mostly in rodents. The reasons for this are manifold, but in part they center on practical aspects such as availability and cost, compounded by ethical considerations. Mice and rats are most frequently used, and many diseases have been modeled in these species. One question that has of late received more attention is that of species variability. Even if the disease is modeled well in the animal, variability in the drug target between humans and rodents may still lead to failed human trials, a venture that is both costly and time-consuming. One example of this is 12/15-lipoxygenase (12/15-LOX), an enzyme, which contributes to ischemic brain injury in both humans and mice.
100051 The lipoxygenases form a large family of enzymes capable of oxidizing arachidonic acid (AA) and related polyunsaturated fatty acids (Brash AR, J. Biol. Chem.
1999, 274, 23679-23682). In humans, in addition to 12/15-LOX, other members include 5-LOX, P-12-LOX, Date Recue/Date Received 2020-11-17 12(R)-LOX, epidermal LOX-3, and 15-LOX-2. Their nomenclature is based in part upon the tissue where they were first detected, and in part upon the carbon atom in AA
that is oxidized.
Correspondingly, 12/15-LOX can oxidize both C12 and C15, forming 12- or 15-hydroperoxyarachidonic acid (12- or 15-HPETE), respectively. Human lipoxygenases and their metabolites have been implicated in numerous diseases. 5-LOX has been implicated in cancer, asthma, COPD, allergic rhinitis, osteoarthritis, and atherosclerosis, while platelet-type 12-LOX
has been implicated in diabetes, blood coagulation, psoriasis, and cancer.
Human reticulocyte 15-lipoxygenase-1 (12/15-LOX, ak.a 15-LOX-1.) is also an. attractive therapeutic target, particularly for its role in atherogenesis, diabetes, Alzheimer's, newborn periventricular leukomalacia, breast cancer and stroke (Tong, W. G., et al., Clln. Can. Res. 2002, 8, 3232-3242;
Pratico, D.;
Zhukareva, V., et al., Am. J. .Pathol. 2004, /64, 1655-1662).
100061 The 12/15-LOX has historically been called 15-LOX, 15-LOX-1, or 15-LO-I in humans, L-I2-LOX, leukocyte-type 12-LO, or L-12-LO in mice. The number prefix reflects the preference for 15-HETE in humans, vs. mostly 12-HETE for the mouse 12/15-LOX.
The gene in both organisms is termed ALOX15, reflecting the close homology (around 78%) and functional equivalence in both species. Mutation of a key residue in the active site of the rabbit 12/15-LOX
which normally generates mostly 15-HETE, switches the product ratio towards the 12 product (Borngraber et al., J. Biol. Chem. 1999, 274, 37345-37350). The variability in the product spectrum. thus reflects differences in the active site binding pocket, which is otherwise similar in most lipoxygenases, although the orientation of arachidonic acid presumably varies (Schwarz et al., Biochemistry 1998, 37, 15327-15335).
[0007] The difficulty in developing inhibitors that target LOX homologues in both species is that they have different substrate specificities and presumably different inhibitor specificities.
Thus, existing inhibitors of 12/15-LOX are typically not very selective with regard to other LOX
isoforms, and many additionally are strong antioxidants. While this latter fact may not in itself be damaging, the inherent lack of selectivity suggests a much greater potential for off-target effects.
100081 Introducing humanized versions of the drug target into the animal model is one way to meet the challenge of species-to-species variability, but this approach is in most cases not available. It is therefore of great importance to identify bioactive compounds that target a specific pathway both in rodents, and in humans.
2 SUMMARY
[00091 Provided herein are compositions and methods for treating a condition involving 12/15-lipoxygenase. Through a systematic succession of screening steps, the inventors have identified novel inhibitors of human 12/I 5-lipoxygenase (12/15-1,0X), which can also target the mouse 12/15-LOX homologue.
[00101 In one aspect, provided herein is a method of treating a condition involving 12/15-lipoxygenase in a subject, the method comprising administering to the subject an effective amount of a compound of Formula I:
R.12 X N

R11 ________________________ <\\
N
Formula I
or a pharmaceutically acceptable salt thereof, in which:
X is 0 or S;
R" is an aryl, heteroaryl, cyclyl, or heterocyclyl, each of which can be optionally substituted;
and R12 and R13 are independently hydrogen, halogen, alkyl, alkenyl, allcynyl, aralkyl, acyl, aryl, heteroaryl, cyclyi or heterocyclyl, each of which can be optionally substituted.
110011] In some embodiments, is an aryl or heteroaryl, each of which can be optionally substituted.
[00121 In some embodiments, R" is 1-naphthyl, 2-naphthyl, 6-isoquinolinyl, 2,3-dichlorophenyl, or 3,4-dichlorophenyl.
[0013] In some embodiments, R12 is hydrogen, alkyl, aralkyl, acyl, aryl, or heterocyclyl, each of which can be optionally substituted.
[0014] In some embodiments, R12 is hydrogen, methyl, ethyl, propyl, butyl, pentyl, or phenyl.
[00151 In some embodiments, R13 is hydrogen, alkyl, aralkyl, acyl, aryl, or heterocyclyl, each of which can be optionally substituted.
3 [00161 In some embodiments, R13 is hydrogen, methyl, ethyl, propyl, butyl, pentyl, or phenyl.
[00171 In some embodiments, RI2 is hydrogen and RI3 is hydrogen, methyl, ethyl, propyl, butyl, pentyl, or phenyl.
[00181 In some embodiments, X is 0.
[00191 In some embodiments, X is S.
[00201 In some embodiments, the compound of Formula I is of:
(i) Formula H:

X
i_Cr NH
CN
Formula II
or a pharmaceutically acceptable salt thereof, in which:
X is 0 or S; and R' is an aryl, heteroaryl, cyclyl, or heterocyclyl, each of which can be optionally substituted; or (ii) Formula III:
-\\ R12 () CN
Formula III
or a pharmaceutically acceptable salt thereof, in which:
R12 and R13 are independently hydrogen, halogen, alkyl, alkenyl, alkynyl, aralkyl, acyl, aryl, heteroaryl, cycly1 or heterocyclyl, each of which can be optionally substituted.
4 [00211 In some embodiments, the compound is selected from the group consisting of 5-(methylamino)-2-naphthalen- 1 -yl- 1 ,3-oxazole-4-carbonitrile (ML351), 2-(2,3-dichlorophenyD-5-(methylamino)- 1 ,3-oxazole-4-carbonitrile, 2-(3,4-dichloropheny1)-5-(methylamino)-1,3-oxazole-4-carbonitri le, 5-(methylamino)-2-naphtha len-1 -y1- 1 ,3-thiazole-4-carbonitri le.
[00221 In some embodiments, the compound is 5-(methylamino)-2-naphthalen-1 -y1- 1 ,3-oxazole-4-carbonitrilc (ML351).
[00231 In some embodiments, the compound inhibits 12/15-lipoxygenase.
[00241 In some embodiments, the condition is stroke, periventricular leukomalacia, cardiac arrest with resuscitation, atherosclerosis. Parkinson's disease, Alzheimer's disease, or breast cancer.
[00251 in some embodiments, the subject is a mammal.
100261 In some embodiments, the subject is a human.
100271 In another aspect, provided herein is the use of a compound of Formula I:

N
X
R11 __ ,<\
N
CN
Formula 1 or a pharmaceutically acceptable salt thereof for the preparation of a medicament for the treatment of a condition involving 12/15-lipoxygenase, wherein:
X is 0 or S;
R11 is an aryl, heteroaryl, cyclyl, or heterocyclyl, each of which can be optionally substituted;
and R12 and R13 are independently hydrogen, halogen, alkyl, alkenyl, alkynyl, aralkyl, acyl, aryl, heteroaryl, cyclyl or heterocyclyl, each of which can be optionally substituted.
[00281 In some embodiments, the compound of Formula I is of:
(i) Formula II:

CH:, X I
Ri __________________________ N-----Z----C N
Formula 11 or a pharmaceutically acceptable salt thereof, wherein:
X is 0 or S; and Rn is an aryl, heteroaryl, cyelyl, or heterocyclyl, each of which can be optionally substituted; or (ii) Formula III:
--------\ / I
"....R.13 111 \
0-............./N

N''''........
CN
Formula 111 or a pharmaceutically acceptable salt thereof, wherein:
RI2 and RI3 are independently hydrogen, halogen, alkyl, alkenyl, alkynyl, aralkyl, acyl, aryl, heteroaryl, cyclyl or heterocyclyl, each of which can be optionally substituted.
[00291 In some embodiments, the compound is selected from the group consisting of 5-(methylamino)-2-naphthalen-l-y1-1,3-oxazole-4-carbonitrile (MI,351), 2-(2,3-dichloropheny1)-5-(methylamino)-1,3-oxazole-4-carbonitrile, 2-(3,4-dichloropheny1)-5-(methylamino)-1,3-oxazole-4-carbonitrile, and 5-(methylamino)-2-naphthalen-l-y1-1,3-thiazole-4-carbonitrile.
[00301 In some embodiments, the condition is stroke, periventricular leukomalacia, cardiac arrest with resuscitation, atherosclerosis, Parkinson's disease, Alzheimer's disease, or breast cancer.
BRIEF DESCRIPTION OF THE DRAWINGS
100311 FIGs. 1A-1J are synthesis schemes of the various compounds described in the present invention.

[00321 FIGs. 2A-2B show correlation between arachidonic acid metabolites and cell death.
[00331 FIG. 2A is a graph showing % of the 12-HETE secreted from glutamate-treated cells vs % cell death in glutamate-treated samples.
[00341 FIG. 2B is a graph showing positive correlation between 15-HETE and cell death.
[00351 FIG. 3A is a plot showing protection against glutamate-induced HT-22 death by increasing amounts of 1 (** p <0.01, *** P <0.001 vs glutamate only).
[00361 FIG. 3B is a plot showing inhibition of 12-HETE in HT-22 cells by 10 uM of 1, following treatment with 5 mM glutamate (* P <0.05 vs glutamate only).
[00371 FIG. 4 is a plot showing cellular protection at 5 LtM concentration in HT22 cells by 1 and some analogs. Despite not inhibiting human 12/15-LOX, 1.1. and 31 show similar protection to 1, indicating they inhibit the mouse enzyme, while 34 does not protect.
[00381 FIG. SA is a plot showing activity of compound 1(50 mg/kg) administered via intraperitoneal. (IP) injection in mouse distal MC.A0 model of permanent focal ischemia (** p <
0.01).
[00391 FIG. 5B is a set of images showing typical examples of sequential (top to bottom represents front to back sections of a single brain) TIC-stained brain sections from. vehicle- and Compound 1-treated mice. The white areas in the cortex (top right) indicate non-viable infarcted tissue.
[00401 FIGs. 6A-6D are plots showing stability of 1 measured as percent composition of probe molecule in aqueous solution (contains 20 A) acetonitrile) at r.t. over 48 hr in (FIG. 6A) DPBS buffer (pH 7.4), (FIG. 6B) Lipoxygenase UV-Vis assay buffer (1M HEPES
buffer, pH
7.3), (FIG. 6C) pH 2 buffer, and (FIG. 6D) pH 10 buffer.
[00411 FIGs. 7A-7B are plots showing steady-state kinetics data for the determination of Ki and Ki`for 12/15-LOX with 1: (FIG. 7A) Plot of 144Nmax versus inhibitor concentration ( M) yielded a Ki of 0.1 0.002 M. (FIG. 7B) Plot of 1/V.versus inhibitor concentration (pM) yielded a IS' of 1.2 0.02 uM (Vm units are timol/minimg).
[00421 FIG. 8 is a dose response curves for probe ML351 showing inhibition of 15-LOX-1 in the UV-Vis assay.
[00431 FIG. 9A is a plot showing that hemorrhage in the ischernic hemisphere of warfarin-treated mice is significantly reduced by ML351. **p<0.001 WO 2015/027146 PC1'11152014/052269 [00441 FIG. 9B is a plot showing that hemorrhage in the ischemic hemisphere of warfarin-treated mice subjected to experimental stroke and treated with tPA is also significantly reduced by MI.351. ***p<0.0001 DETAILED DESCRIPTION
[00451 Through a systematic succession of screening steps, the inventors have identified novel inhibitors of human 12/15-lipoxygenase 2/15-1.,OX), which can also target the mouse 12/15-LOX homologue. Accordingly, embodiments of the present invention provide, inter alia, 12115-LOX inhibitors that work against both the human and the mouse version of and methods of uses thereof.
[00461 One aspect of the invention relates to small molecule compounds that are 12/15-LOX
inhibitors. In some embodiments, the compound corresponds to Formula I:

X
R" _________________________ Formula 1 or a pharmaceutically acceptable salt thereof.
[00471 In some embodiments, the compound of Formula us of (i) Formula II:

X
R11 ____________________________________ NH
CN
Formula II
or a pharmaceutically acceptable salt thereof; or (ii) Formula III:

N"-R13 Formula III
or a pharnriaceutically acceptable salt thereof.
10048j In compounds of Formula I or II, X can be 0 or S. and RH can be an aryl, heteroaryl, cyclyl, or heterocyclyl, each of which can be optionally substituted.
100491 In compounds of Formula I or III, R12 and R13 are independently hydrogen, halogen, alkyl, alkenyl, alkynyl, aralkyl, acyl, aryl, heteroaryl, cyclyl or heterocyclyl, each of which can be optionally substituted.
[00501 In some embodiments, RH is an aryl or heteroaryl, each of which can be optionally substituted. In some embodiments, R" is a monocyclic aryl, bicyclic aryl, monocyclic heteroaryl, or bicyclic heteroaryl, each of which can be optionally substituted. In some embodiments, R" is an optionally substituted phenyl. When the phenyl group is substituted, the substituent can be present at the ortho, meta, or para position on the phenyl relative to the rest of the compound. In some embodiments, the optionally substituted aryl or heteroaryl is substituted with one or more substitucnts selected from amino, halogen, hydroxyl, thiol, methoxy, methylthioxy, carboxyl, nitro, cyano, and any combinations thereof. In some embodiments, the heteroaryl comprises one or more heteroatoms (e.g., 1, 2, 3, 4, 5, or more) selected from 0, S, N, and a combination thereof.
[0051] In some embodiments, R11 is 1-naphthyl, 2-naphthyl, 5-isoquinolinyl, 6-isoquinolinyl,
5-quinolinyl, 2,3-dichlorophenyl, 3,4-dichlorophenyl, 5-benzo[d][1,3]dioxolyl, 3-1H-indolyl, or 3-chlorophenyl. In some embodiments, RH is 1-naphthyl, 2-naphthyl, 6-isoquinolinyl, 2,3-dichlorophenyl, or 3,4-dichlorophenyl.
[0052] In some embodiments, R12 and R13 are independently hydrogen, alkyl, aralkyl, alkenyl, acyl, aryl, or heterocyclyl, each of which can be optionally substituted. In some embodiments, the alkyl is C1-Cio alkyl. In some embodiments, the alkyl is CI-C6 alkyl such as
6 PCTIUS2014/052269 methyl, ethyl, propyl, isopropyl, butyl, sec-butyl, isobutyl, tert-butyl, pentyl, neopentyl, and hexyl.
[00531 In some embodiments, when R12 is hydrogen, R13 can be methyl, ethyl, propyl, butyl, pentyl, formyl, acetyl, benzyl, or phenyl, [00541 In some embodiments, R12 and R13 are both methyl.
[00551 In some embodiments, the compound is selected from the group consisting of 5-(methylamino)-2-naphthalen- I -y1-1,3-oxazole-4-carbonitrile (also referred herein as ML351), 2-(2,3-diehlorophenyI)-5-(methylamino)-1,3-oxazole-4-carbonitrile, 2-(3,4-dichloropheny1)-5-(methylamino)-1,3-oxazole-4-carbonitrile, 5-(methylamino)-2-naphthalen-l-y1-1,3-thiazole-4-carbonitrile.
(00561 In some embodiments, the compound is 5-(methylamino)-2-naphthalen-l-y1-1,3-oxazole-4-carbonitrile (ML351).
100571 In some embodiments, the compound can cross the blood-brain barrier.
(00581 In some embodiments, the compound can selectively inhibit 12/15-LOX.
100591 Set forth below are some specific examples of the compounds which can be used to practice the methods of this invention:

NH H
\ I
Compound I Compound 2 NH N/
NH
"====-\ \ I
CN
Compound 3 Compound 4 NH CI CI
NH
CN
NCN
\
N
Compound 5 Compound 7 CI
CN
\

Compound 8 Compound 10 CI
NH
HN NCN
\ \
Compound 14 Compound 16 NH
\ I
Compound 20 Compound 21 NH
\ NCN
Compound 22 Compound 23 \
NCN
NCN
Compound 24 Compound 25 11 Oy NH 411 .

\
N
\\\\, ___________________________________ ty CN N"------CN
Compound 27 Compound 2 / \ 401 /...... .....
/ \
NH
NH S-----.."
0-..............","
\ Ilk \
N-----CN
N'....-..'%.***=,CN
Compound 32 Compound 29 I
, RI Fi y c.
------ \
Q. NH
0-........õ.õ...-KN\ __ , __ ( IN
N
Compound 35 Compound 38 100601 Compounds of Formula 1, 11, or III are either commercially available or can be synthesized by methods well known to one of skill in the chemical arts.
Exemplary methods to synthesize compound 1 and other compounds can be found in the Examples section.
10061] Methods for measuring the inhibitory effect of a compound on 12/15-LOX are also well known in the art. For example, lipoxygenase UV-Vis assays can be used.
The lipoxygenase 1N-Vis assay can permit the determination of percent inhibition and 1050 values. 1050, also called the half maximal inhibitory concentration, is a measure of the effectiveness of a substance in inhibiting a specific biological or biochemical function. IC50 values can be obtained, for example, by determining the enzymatic rate at various inhibitor concentrations and plotted against inhibitor concentration, followed by a hyperbolic saturation curve fit.
Pharmaceutical compositions [00621 Another aspect of the invention relates to pharmaceutical compositions comprising the compounds (i.e., 12/15-LOX inhibitors) disclosed herein. In some embodiments, the pharmaceutical composition comprises a pharmaceutically-acceptable carrier and/or diluent.
Some examples of materials which can serve as pharmaceutically-acceptable carriers include:
(1) sugars, such as lactose, glucose and sucrose; (2) starches, such as corn starch and potato starch; (3) cellulose, and its derivatives, such as sodium. carboxymethyl cellulose, methylcellulose, ethyl cellulose, microcrystalline cellulose and cellulose acetate; (4) powdered tragacanth; (5) malt; (6) gelatin; (7) lubricating agents, such as magnesium stearate, sodium lauryl sulfate and talc; (8) excipients, such as cocoa butter and suppository waxes; (9) oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; (10) glycols, such as propylene glycol; (11) polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol; (12) esters, such as ethyl ol.eate and ethyl laurate;
(13) agar; (14) buffering agents, such as magnesium. hydroxide and aluminum hydroxide; (15) alginic acid; (16) pyrogen-free water; (17) isotonic saline; (18) Ringer's solution; (19) ethyl alcohol;
(20) pH buffered solutions; (21) polyesters, polycarbonates and/or polyanhydrides; (22) bulking agents, such as polypeptides and amino acids (23) serum component, such as serum albumin, HDL
and LDL;
(22) C2-C12 alcohols, such as ethanol; and (23) other non-toxic compatible substances employed in pharmaceutical formulations. Wetting agents, coloring agents, release agents, coating agents, sweetening agents, flavoring agents, perfuming agents, preservative and antioxidants can also be present in the formulation. The terms such as "excipient", "carrier", "pharmaceutically acceptable carrier" or the like are used interchangeably herein. The amount of a 12/15-LOX
inhibitor which can be combined with a carrier material to produce a single dosage form will generally be that amount of the 12/15-LOX inhibitor which produces a desired therapeutic effect.
Generally out of one hundred percent, this amount will range from about 0.01%
to 99% of 12/15-LOX inhibitor, such as from about 0.1% to about 70%, and from 5% to about 30%.

[00631 Additionally, the 12/15-LOX inhibitors can be delivered using lipid-or polymer-based nanoparticles. See for example Allen, T.M., Cullis, P.R. Drug delivery systems: entering the mainstream. Science. 303(5665): 1818-22 (2004).
[00641 in some embodiments, the pharmaceutical compositions further comprise an agent (e.g., a vehicle) that facilitates crossing the blood-brain barrier by the 12/15-LOX inhibitors.
100651 The pharmaceutical compositions of the present invention can be specially formulated for administration in solid, liquid or gel form, including those adapted for the following: (1) oral administration, for example, drenches (aqueous or non-aqueous solutions or suspensions), lozenges, drage, capsules, pills, tablets (e.g., those targeted for buccal, sublingual, and systemic absorption), boluses, powders, granules, pastes for application to the tongue; (2) parenteral administration, for example, by subcutaneous, intramuscular, intravenous or epidural injection as, for example, a sterile solution or suspension, or sustained-release formulation; (3) topical. application, for example, as a cream, ointment, or a controlled-release patch or spray applied to the skin; (4) intravaginally or intrarectally, for example, as a pessary, cream or foam;
(5) sublingually; (6) ocularly; (7) transdermally; (8) transmucosally; or (9) nasally. Additionally the compounds described herein can be implanted into a patient or injected using a drug delivery system. See, for example, Urquhart, etal., Ann. Rev. Phammeol. Toxicol. 24:
199-236 (1984);
Lewis, ed. "Controlled Release of Pesticides and Pharmaceuticals" (Plenum Press, New York, 1981); U.S. Pat. No. 3,773,919; and U.S. Pat. No. 35 3,270,960. Examples of dosage forms include, but are not limited to: tablets; caplets; capsules, such as hard gelatin capsules and soft elastic gelatin capsules; cachets; troches; lozenges; dispersions;
suppositories; ointments;
cataplasms (poultices); pastes; powders; dressings; creams; plasters;
solutions; patches; aerosols (e.g., nasal sprays or inhalers); gels; liquids such as suspensions (e.g., aqueous or non-aqueous liquid suspensions, oil-in-water emulsions, or water-in-oil liquid emulsions.), solutions, and elixirs; and sterile solids (e.g., crystalline or amorphous solids) that can be reconstituted to provide liquid dosage forms.
[00661 Aerosol formulations. 12/15-LOX inhibitors can be administered directly to the airways in the form of an aerosol or by nebulization. For use as aerosols, 12/15-LOX inhibitors in solution or suspension may be packaged in a pressurized aerosol container together with suitable propellants, for example, hydrocarbon propellants like propane, butane, or isobutane with conventional adjuvants. The 12/15-LOX inhibitors can also be administered in a non-pressurized form such as in a nebulizer or atomizer.
[00671 The term "nebulization" is well known in the art to include reducing liquid to a fine spray. Preferably, by such nebulization small liquid droplets of uniform size are produced from a larger body of liquid in a controlled manner. Nebulization can be achieved by any suitable means therefor, including by using many nebulizers known and marketed today. For example, an.
AEROMIST pneumatic nebulizer is available from Inhalation Plastic, Inc. of Niles, Ill.
[00681 As is well known, any suitable gas can be used to apply pressure during the nebulization, with preferred gases to date being those which are chemically inert to the 12/15-LOX inhibitors. Exemplary gases including, but are not limited to, nitrogen, argon or helium can be used to high advantage.
[00691 12/15-LOX inhibitors can also be administered directly to the airways in the form of a dry powder. For use as a dry powder, 12/15-LOX inhibitors can be administered by use of an inhaler. Exemplary inhalers include metered dose inhalers and dry powdered inhalers. A.
metered dose inhaler or "MDI" is a pressure resistant canister or container filled with a product such as a pharmaceutical composition dissolved in a liquefied propellant or micronized particles suspended in a liquefied propellant. The correct dosage of the composition is delivered to the patient. A dry powder inhaler is a system operable with a source of pressurized air to produce dry powder particles of a pharmaceutical composition that is compacted into a very small volume.
100701 Dry powder aerosols for inhalation therapy are generally produced with mean diameters primarily in the range of <5gm. As the diameter of particles exceeds 3gm, there is increasingly less phagocytosis by macrophages. However, increasing the particle size also has been found to minimize the probability of particles (possessing standard mass density) entering the airways and acini due to excessive deposition in the oropharyngeal or nasal regions.
[00711 Suitable powder compositions include, by way of illustration, powdered preparations of 12/15-LOX inhibitors thoroughly intermixed with lactose, or other inert powders acceptable for intrabronchial administration. The powder compositions can be administered via an aerosol dispenser or encased in a breakable capsule which may be inserted by the patient into a device that punctures the capsule and blows the powder out in a steady stream suitable for inhalation.

The compositions can include propellants, surfactants, and co-solvents and may be filled into conventional aerosol containers that are closed by a suitable metering valve.
[00721 Aerosols for the delivery to the respiratory tract are known in the art. See for example, Adjei, A. and Garren, J. Pharm. Res., 1: 565-569 (1990); Zanen, P.
and Lamm, J.-W. J.
Int. J. Pharm., 114: 111-115 (1995); Gonda, 1. "Aerosols for delivery of therapeutic and diagnostic agents to the respiratory tract," in Critical Reviews in Therapeutic Drug Carrier Systems, 6:273-313 (1990); Anderson et al., Am. Rev. Respir. Dis., 140: 1317-1324 (1989)) and have potential for the systemic delivery of peptides and proteins as well (Patton and Platz, Advanced Drug Delivery Reviews, 8:179-196 (1992)); Timsina et. al., Int. J.
Pharm., 101: 1-13 (1995); and Tansey, I. P., Spray Technol. Market, 4:26-29 (1994); French, D.
L., Edwards, D. A.
and Niven, R. W., Aerosol Sci., 27: 769-783 (1996); Visser, J., Powder Technology 58: 1-10 (1989)); Rudt, S. and R. H. Muller, J. Controlled Release, 22: 263-272 (1992);
Tabata, Y, and Y.
Ikada, Biomed. Mater. Res., 22: 837-858 (1988); Wall, D. A., Drug Deliveiy, 2:
10 1-20 1995);
Patton, J. and Platz, R., Adv. Drug Del. Rev., 8: 179-196 (1992); Bryon, P., Adv. Drug. Del.
Rev., 5: 107-132 (1990); Patton, J. S., et al., Controlled Release, 28: 15 79-85 (1994); Damms, B. and Bains, W., Nature Biotechnology (1996); Niven, R. W., et al., Pharm.
Res., 12(9); 1343-1349 (1995); and Kobayashi, S., et al., Pluirm. Res., 13(1): 80-83 (1996).
[00731 Oral formulations. Pharmaceutical compositions comprising 12/15-LOX
inhibitors of the present invention can also be formulated into oral dosage forms such as, but not limited to, tablets (including without limitation scored or coated tablets), pills, caplets, capsules, chewable tablets, powder packets, cachets, troches, wafers, aerosol sprays, or liquids, such as but not limited to, syrups, elixirs, solutions or suspensions in an aqueous liquid, a non-aqueous liquid, an oil-in-water emulsion, or a water-in-oil emulsion. Such compositions contain a predetermined amount of the pharmaceutically acceptable salt of the disclosed compounds, and may be prepared by methods of pharmacy well known to those skilled in the art. See generally, Remington's Pharmaceutical Sciences, 18th ed., Mack Publishing, Easton, Pa.
(1990).
[00741 Typical oral dosage forms are prepared by combining the pharmaceutically acceptable salt of the disclosed compounds in an intimate admixture with at least one excipient according to conventional pharmaceutical compounding techniques. Excipients can take a wide variety of forms depending on the form of the composition desired for administration. For Date Recue/Date Received 2020-11-17 example, excipients suitable for use in oral liquid or aerosol dosage forms include, but are not limited to, water, glycols, oils, alcohols, flavoring agents, preservatives, and coloring agents.
Examples of excipients suitable for use in solid oral dosage forms (e.g., powders, tablets, capsules, and caplets) include, but are not limited to, starches, sugars, microcrystalline cellulose, kaolin, diluents, granulating agents, lubricants, binders, and disintegrating agents. Due to their ease of administration, tablets and capsules represent the most advantageous solid oral dosage unit forms, in which case solid pharmaceutical excipients are used. If desired, tablets can be coated by standard aqueous or nonaqueous techniques. These dosage forms can be prepared by any of the methods of pharmacy. In general, pharmaceutical compositions and dosage forms are prepared by uniformly and intimately admixing the active ingredient(s) with liquid carriers, finely divided solid carriers, or both, and then shaping the product into the desired presentation if necessary.
[00751 For example, a tablet can be prepared by compression or molding.
Compressed tablets can be prepared by compressing in a suitable machine the active ingredient(s) in a free-flowing form, such as a powder or granules, optionally mixed with one or more excipients.
Molded tablets can be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent. Examples of excipients that can be used in oral dosage forms of the disclosure include, but are not limited to, binders, fillers, disintegrants, and lubricants. Binders suitable for use in pharmaceutical compositions and dosage forms include, but are not limited to, corn starch, potato starch, or other starches, gelatin, natural and synthetic gums such as acacia, sodium alginate, alginic acid, other alginates, powdered tragacanth, guar gum, cellulose and its derivatives (e.g., ethyl cellulose, cellulose acetate, carboxymethyl cellulose calcium, sodium carboxymethyl cellulose), polyvinyl pyrrolidone, methyl cellulose, pre-gelatinized starch, hydroxypropyl methyl cellulose, microcrystalline cellulose, and mixtures thereof.
[00761 Suitable forms of microcrystalline cellulose include, but are not limited to, the materials sold as AV10EL-PH-101 , AV10EL-PH-103 AViCEL RC-581 , and AVICEL- PH-105 (available from FMC Corporation, American Viscose Division, Avicel Sales, Marcus Hook, Pa., U.S.A.), and mixtures thereof. An exemplary suitable binder is a mixture of microcrystalline cellulose and sodium carboxymethyl cellulose sold as AV10EL RC-581. Suitable anhydrous or low moisture excipients or additives include AVICEL-PH-103"1 and Starch 1500 LM.

[00771 Examples of fillers suitable for use in the pharmaceutical compositions and dosage forms disclosed herein include, but are not limited to, talc, calcium carbonate (e.g., granules or powder), microcrystalline cellulose, powdered cellulose, dextrates, kaolin, mannitol, silicic acid, sorbitol, starch, pre-gelatinized starch, and mixtures thereof. The binder or filler in pharmaceutical compositions of the disclosure is typically present in from about 50 to about 99 weight percent of the pharmaceutical composition or dosage form.
[00781 Disintegrants are used in the compositions of the disclosure to provide tablets that disintegrate when exposed to an aqueous environment. Tablets that contain too much disintegrant may swell, crack, or disintegrate in storage, while those that contain too little may be insufficient for disintegration to occur and may thus alter the rate and extent of release of the active ingredient(s) from. the dosage form. Thus, a sufficient am.ount of disintegrant that is neither too little nor too much to detrimentally alter the release of the active ingredient(s) should be used to form solid oral dosage forms of the disclosure. The amount of disintegrant used varies based upon the type of formulation and mode of administration, and is readily discernible to those of ordinary skill in the art. Typical pharmaceutical compositions comprise from about 0.5 to about 15 weight percent of disintegrant, preferably from about 1. to about 5 weight percent of disintegrant.
[00791 Disintegrants that can be used to form pharmaceutical compositions and dosage forms of the disclosure include, but are not limited to, agar, alginic acid, calcium carbonate, microcrystalline cellulose, croscarmellose sodium, crospovidone, polacrilin potassium, sodium starch glycolate, potato or tapioca starch, other starches, pre-gelatinized starch, clays, other algins, other celluloses, gums, and mixtures thereof.
100801 Lubricants that can be used to form pharmaceutical compositions and dosage forms of the disclosure include, but are not limited to, calcium stearate, magnesium stearate, mineral oil, light mineral oil, glycerin, sorbitol, mannitol, polyethylene glycol, other glycols, stearic acid, sodium lauryl sulfate, talc, hydrogenated vegetable oil (e.g., peanut oil, cottonseed oil, sunflower oil, sesame oil, olive oil, corn oil, and soybean oil), zinc stearate, ethyl oleate, ethyl laureate, agar, and mixtures thereof. Additional lubricants include, for example, a syloid silica gel (AEROSIL* 200, manufactured by W. R. Grace Co. of Baltimore, Md.), a coagulated aerosol of synthetic silica (marketed by Degussa Co. of Piano, Tex.), CAB-0-SIO (a pyrogenic silicon dioxide product sold by Cabot Co. of Boston, Mass.), and mixtures thereof. If used at all, lubricants are typically used in an amount of less than about I weight percent of the pharmaceutical compositions or dosage forms into which they are incorporated.
[0081] This disclosure further encompasses anhydrous pharmaceutical compositions and dosage forms comprising the disclosed compounds as active ingredients, since water can facilitate the degradation of some compounds. For example, the addition of water (e.g., 5%) is widely accepted in the pharmaceutical arts as a means of simulating long-term storage in. order to determine characteristics such as shelf life or the stability of formulations over time. See, e.g., Jens T. Carstensen, Drug Stability: Principles & Practice, 379-80 (2nd ed., Marcel Dekker, NY, N.Y.: 1995). Water and heat accelerate the decomposition of som.e compounds.
Thus, the effect of water on a formulation can be of great significance since moisture and/or humidity are commonly encountered during manufacture, handling, packaging, storage, shipment, and use of formulations.
[0082] Anhydrous pharmaceutical compositions and dosage forms of the disclosure can be prepared using anhydrous or low moisture containing ingredients and low moisture or low humidity conditions. Pharmaceutical compositions and dosage forms that comprise lactose and at least one active ingredient that comprises a primary or secondary amine are preferably anhydrous if substantial contact with moisture and/or humidity during manufacturing, packaging, and/or storage is expected.
[0083] An anhydrous pharmaceutical composition should be prepared and stored such that its anhydrous nature is maintained. Accordingly, anhydrous compositions are preferably packaged using materials known to prevent exposure to water such that they can be included in suitable formulary kits. Examples of suitable packaging include, but are not limited to, hermetically sealed foils, plastics, unit dose containers (e.g., vials) with or without desiccants, blister packs, and strip packs.
[0084] Controlled and delayed release formulations. 12/15-LOX inhibitors can be administered by controlled- or delayed-release means. Controlled-release pharmaceutical products have a common goal of improving drug therapy over that achieved by their non-controlled release counterparts. Ideally, the use of an optimally designed controlled-release preparation in medical treatment is characterized by a minimum of drug substance being employed to cure or control the condition in a minimum amount of time.
Advantages of controlled-release formulations include: 1) extended activity of the drug; 2) reduced dosage frequency; 3) increased patient compliance; 4) usage of less total drug; 5) reduction in local or systemic side effects; 6) minimization of drug accumulation; 7) reduction in blood level fluctuations; 8) improvement in efficacy of treatment; 9) reduction of potentiation or loss of drug activity; and 10) improvement in speed of control of diseases or conditions.
Kim, Cherng-ju, Controlled Release Dosage Form Design, 2 (Technomie Publishing, Lancaster, Pa.: 2000).
[00851 Conventional dosage forms generally provide rapid or immediate drug release from the formulation. Depending on the pharmacology and pharmacokinetics of the drug, use of conventional dosage forms can lead to wide fluctuations in the concentrations of the drug in a patient's blood and other tissues. These fluctuations can impact a number of parameters, such as dose frequency, onset of action, duration of efficacy, maintenance of therapeutic blood levels, toxicity, side effects, and the like. Advantageously, controlled-release formulations can be used to control a drug's onset of action, duration of action, plasma levels within the therapeutic window, and peak blood levels. In particular, controlled- or extended-release dosage forms or formulations can be used to ensure that the maximum effectiveness of a drug is achieved while minimizing potential adverse effects and safety concerns, which can occur both from under-dosing a drug (i.e., going below the minimum therapeutic levels) as well as exceeding the toxicity level for the drug.
100861 Most controlled-release formulations are designed to initially release an amount of drug (active ingredient) that promptly produces the desired therapeutic effect, and gradually and continually release other amounts of drug to maintain this level of therapeutic or prophylactic effect over an extended period of time. In order to maintain this constant level of drug in the body, the drug must be released from the dosage form at a rate that will replace the amount of drug being metabolized and excreted from the body. Controlled-release of an active ingredient can be stimulated by various conditions including, but not limited to, pH, ionic strength, osmotic pressure, temperature, enzymes, water, and other physiological conditions or compounds.
[00871 A variety of known controlled- or extended-release dosage forms, formulations, and devices can be adapted for use with the salts and compositions of the disclosure. Examples include, but are not limited to, those described in U.S. Pat. Nos.: 3,845,770;
3,916,899;
3,536,809; 3,598,123; 4,008,719; 5674,533; 5,059,595; 5,591 ,767; 5,120,548;
5,073,543;
5,639,476; 5,354,556; 5,733,566; and 6,365,185 Bl.
These dosage forms can be used to provide slow or controlled-release of one or more Date Recue/Date Received 2020-11-17 active ingredients using, for example, hydroxypropylmethyl cellulose, other polymer matrices, gels, permeable membranes, osmotic systems (such as OROS (Alza Corporation, Mountain View, Calif. USA)), multilayer coatings, microparticles. Liposomes, or microspheres or a combination thereof to provide the desired release profile in varying proportions. Additionally, ion exchange materials can be used to prepare immobilized, adsorbed salt forms of the disclosed compounds and thus effect controlled delivery of the drug. Examples of specific anion exchangers include, but are not limited to, Duolite A568 and Duolite AP143 (Rohm&Haas, Spring House, Pa. USA).
[0088] One embodiment of the disclosure encompasses a unit dosage form that includes a pharmaceutically acceptable salt of the disclosed compounds (e.g., a sodium, potassium, or lithium salt), or a poirtiorph, solvate, hydrate, dehydrate, co- crystal, anhydrous, or amorphous form thereof, and one or more pharmaceutically acceptable excipients or diluents, wherein the pharmaceutical composition or dosage form is formulated for controlled-release. Specific dosage forms utilize an osmotic drug delivery system.
[0089] A particular and well-known osmotic drug delivery system is referred to as OROS
(Alza Corporation, Mountain View, Calif. USA). This technology can readily be adapted for the delivery of compounds and compositions of the disclosure. Various aspects of the technology are disclosed in U.S. Pat. Nos. 6,375,978 B1 ; 6,368,626 B1; 6,342,249 B1 ;
6,333,050 B2;
6,287,295 Bl ; 6,283,953 B1 ; 6,270,787 B1 ; 6,245,357 B 1 ; and 6,132,420.
Specific adaptations of OROSI that can be used to administer compounds and compositions of the disclosure include, but are not limited to, the OROS Push-Pull', Delayed PushPuilTM, Multi-Layer PushPullTM, and Push-Stickm Systems, all of which are well known. See, e.g. worldwide website alza.com. Additional OROS systems that can be used for the controlled oral delivery of compounds and compositions of the disclosure include OROS -CT and L-OROS ; see, Delivery Times, vol. 11, issue H (Alza Corporation).
[0090] Conventional OROS oral dosage forms are made by compressing a drug powder (e.g., a 12/15-LOX inhibitor salt) into a hard tablet, coating the tablet with cellulose derivatives to form a semi-permeable membrane, and then drilling an orifice in the coating (e.g., with a laser). Kim, Chemg-ju, Controlled Release Dosage Form Design, 231-238 (Technomic Publishing, Lancaster, Pa.: 2000). The advantage of such dosage forms is that the delivery rate of the drug is not influenced by physiological or experimental conditions. Even a drug with a pH-Date Recue/Date Received 2020-11-17 dependent solubility can be delivered at a constant rate regardless of the pH
of the delivery medium. But because these advantages are provided by a build-up of osmotic pressure within the dosage form after administration, conventional OROS''' drug delivery systems cannot be used to effectively delivery drugs with low water solubility.
[0091] In some embodiments, the 12/15-LOX inhibitor is administered to a subject by sustained release or in pulses. Pulse therapy is not a form of discontinuous administration of the same amount of a composition over time, but comprises administration of the same dose of the composition at a reduced frequency or administration of reduced doses.
Sustained release or pulse administration are particularly preferred when the angiogenesis is associated with tumor growth as it leads to regression of blood vessels feeding the tumor and ultimately to regression of the tumor itself. Each pulse dose can be reduced and the total amount of drug administered over the course of treatment to the patient is minimized.
[0092] The number of pulses in a single therapeutic regimen may be as little as two, but is typically from about 5 to 10, 10 to 20, 15 to 30 or more. In fact, patients can receive drugs for life according to the methods of this invention without the problems and inconveniences associated with current therapies. Compositions can be administered by most any means, but are preferable delivered to the patient as an injection (e.g. intravenous, subcutaneous, intraarterial), infusion or instillation. Various methods and apparatus for pulsing compositions by infusion or other forms of delivery to the patient are disclosed in U.S. Pat. Nos.
4,747,825; 4,723,958;
4,948,592; 4,965,251 and 5,403,590.
[0093] Parenteral formulations. Parenteral dosage forms can be administered to patients by various routes, including, but not limited to, subcutaneous, intravenous (including bolus injection), intramuscular, and intraarterial. Since administration of parenteral dosage forms typically bypasses the patient's natural defenses against contaminants, parenteral dosage forms are preferably sterile or capable of being sterilized prior to administration to a patient. Examples of parenteral dosage forms include, but are not limited to, solutions ready for injection, dry products ready to be dissolved or suspended in a pharmaceutically acceptable vehicle for injection, suspensions ready for injection, and emulsions. In addition, controlled-release parenteral dosage forms can be prepared for administration of a patient, including, but not limited to, administration DUROS1)-type dosage forms, and dose-dumping.

[00941 Suitable vehicles that can be used to provide parenteral dosage forms of the disclosure are well known to those skilled in the art. Examples include, without limitation: sterile water; water for injection USP; saline solution; glucose solution; aqueous vehicles such as but not limited to, sodium chloride injection, Ringer's injection, dextrose Injection, dextrose and sodium chloride injection, and lactated Ringer's injection; water-miscible vehicles such as, but not limited to, ethyl alcohol, polyethylene glycol, and propylene glycol; and non-aqueous vehicles such as, but not limited to, corn oil, cottonseed oil, peanut oil, sesame oil, ethyl oleate, isopropyl myristate, and benzyl benzoate.
[00951 Compounds that alter or modify the solubility of a pharmaceutically acceptable salt of a 12/15-LOX inhibitor disclosed herein can also be incorporated into the parenteral dosage forms of the disclosure, including conventional and controlled-release parenteral dosage forms.
[00961 Topical, transdermal, and mucosa] formulations. Topical dosage forms of the disclosure include, but are not limited to, creams, lotions, ointments, gels, shampoos, sprays, aerosols, solutions, emulsions, and other forms known to one of skill in the art. See, e.g., Remington's Pharmaceutical Sciences, 18th ed., Mack Publishing, Easton, Pa.
(1990); and Introduction to Pharmaceutical Dosage Forms, 4th ed., Lea & Febiger, Philadelphia, Pa. (1985).
For non-sprayable topical dosage forms, viscous to semi-solid or solid forms comprising a carrier or one or more excipients compatible with topical application and having a dynamic viscosity preferably greater than water are typically employed. Suitable formulations include, without limitation, solutions, suspensions, emulsions, creams, ointments, powders, liniments, salves, and the like, which are, if desired, sterilized or mixed with auxiliary agents (e.g., preservatives, stabilizers, wetting agents, buffers, or salts) for influencing various properties, such as, for example, osmotic pressure. Other suitable topical dosage forms include sprayable aerosol preparations wherein the active ingredient, preferably in combination with a solid or liquid inert carrier, is packaged in a mixture with a pressurized volatile (e.g., a gaseous propellant, such as freon), or in a squeeze bottle. Moisturizers or humectants can also be added to pharmaceutical compositions and dosage forms if desired. Examples of such additional ingredients are well known in the art. See, e.g., Remington's Pharmaceutical Sciences, 18th Ed., Mack Publishing, Easton, Pa. (1990).
[00971 Transdermal and mucosal dosage forms of the 12/15-LOX inhibitor compositions of the disclosure include, but are not limited to, ophthalmic solutions, patches, sprays, aerosols, creams, lotions, suppositories, ointments, gels, solutions, emulsions, suspensions, or other forms known to one of skill in the art. Dosage forms suitable for treating mucosal tissues within the oral cavity can be formulated as mouthwashes, as oral gels, or as buccal patches. Additional transdermal dosage forms include "reservoir type" or "matrix type" patches, which can be applied to the skin and worn for a specific period of time to permit the penetration of a desired amount of active ingredient.
[00981 Examples of transdermal dosage forms and methods of administration that can be used to administer the active ingredient(s) of the disclosure include, but are not limited to, those disclosed in U.S. Pat. Nos.: 4,624,665; 4,655,767; 4,687,481; 4,797,284;
4,810,499; 4,834,978;
4,877,618; 4,880,633; 4,917,895; 4,927,687; 4,956,171 ; 5,035,894; 5,091 ,186;
5,163,899;
5,232,702; 5,234,690; 5,273,755; 5,273,756; 5,308,625; 5,356,632; 5,358,715;
5,372,579; 5,421 ,816; 5,466;465; 5,494,680; 5,505,958; 5,554,381; 5,560,922; 5,585,111 ;
5,656,285; 5,667,798;
5,698,217; 5,741 ,511 ; 5,747,783; 5,770,219; 5,814,599; 5,817,332; 5,833,647;
5,879,322; and 5,906,830.
[00991 Suitable excipients (e.g., carriers and diluents) and other materials that can be used to provide transdermal and mucosal dosage forms encompassed by this disclosure are well known to those skilled in the pharmaceutical arts, and depend on the particular tissue or organ to which a given pharmaceutical composition or dosage form will be applied. With that fact in mind, typical excipients include, but are not limited to water, acetone, ethanol, ethylene glycol, propylene glycol, butane-1,3-diol, isopropyl myristate, isopropyl palmitate, mineral oil, and mixtures thereof, to form dosage forms that are non-toxic and pharmaceutically acceptable.
[001001 Depending on the specific tissue to be treated, additional components may be used prior to, in conjunction with, or subsequent to treatment with a 12/15-LOX
inhibitor of the disclosure. For example, penetration enhancers can be used to assist in delivering the active ingredients to or across the tissue. Suitable penetration enhancers include, but are not limited to:
acetone; various alcohols such as ethanol, oleyl, an tetrahydrofuryl; alkyl sulfoxides such as dimethyl sulfoxide; dimethyl acetamide; dimethyl formamide; polyethylene glycol; pyrrolidones such as polyvinylpyrrolidone; Kollidon grades (Povidone, Polyvidone); urea;
and various water-soluble or insoluble sugar esters such as TWEEN 80 (polysorbate 80) and SPAN
60 (sorbitan monostearate).
Date Recue/Date Received 2020-11-17 [00101] The pH of a pharmaceutical composition or dosage form, or of the tissue to which the pharmaceutical composition or dosage form is applied, may also be adjusted to improve delivery of the active ingredient(s). Similarly, the polarity of a solvent carrier, its ionic strength, or tonicity can be adjusted to improve delivery. Compounds such as stearates can also be added to pharmaceutical compositions or dosage forms to advantageously alter the hydrophilicity or lipophil.icity of the active ingredient(s) so as to improve delivery. In this regard, stearates can serve as a lipid vehicle for the formulation, as an emulsifying agent or surfactant, and as a delivery- enhancing or penetration-enhancing agent. Different hydrates, dehydrates, co- crystals, solvates, polymorphs, anhydrous, or amorphous forms of the pharmaceutically acceptable salt of a 12/15-LOX inhibitor can be used to further adjust the properties of the resulting composition.
1001021 The data obtained from the cell culture assays and animal studies can be used in formulating a range of dosage for use in humans. The dosage of such compounds lies preferably within a range of circulating concentrations that include the ED50 with little or no toxicity. The dosage may vary within this range depending upon the dosage form employed and the route of administration utilized.
1001031 The therapeutically effective dose can be estimated initially from cell culture assays.
A dose may be formulated in animal models to achieve a circulating plasma concentration range that includes the IC50 (i.e., the concentration of the therapeutic which achieves a half-maximal inhibition of symptoms) as determined in cell culture. Levels in plasma may be measured, for example, by high performance liquid chromatography. The effects of any particular dosage can be monitored by a suitable bioassay.
1001041 The dosage can be determined by one of skill in the art and can also be adjusted by the individual physician in the event of any complication. Typically, the dosage of a composition comprising a 12/15-LOX inhibitor disclosed herein can range from 0.001mg/kg body weight to 5 g/kg body weight. in some embodiments, the dosage range is from 0.001 mg/kg body weight to 1g/kg body weight, from 0.001 mg/kg body weight to 0.5 g/kg body weight, from 0.001 mg/kg body weight to 0.1 g/kg body weight, from 0.001 mg/kg body weight to 50 mg/kg body weight, from 0.001 mg/kg body weight to 25 mg/kg body weight, from 0.001 mg/kg body weight to 10 mg/kg body weight, from 0.001 mg/kg body weight to 5 mg/kg body weight, from 0.001 mg/kg body weight to 1 mg/kg body weight, from 0.001 mg/kg body weight to 0.1 mg/kg body weight, or from 0.001 mg/kg body weight to 0.005 mg/kg body weight.
Alternatively, in some embodiments the dosage range is from 0.1 g/kg body weight to 5 g/kg body weight, from 0.5 g/kg body weight to 5 g/kg body weight, from 1 g/kg body weight to 5 g/kg body weight, from 1.5 g/kg body weight to 5 glkg body weight, from 2 g/kg body weight to 5 g/kg body weight, from 2.5 g/kg body weight to 5 g/kg body weight, from 3 g/kg body weight to 5 g/kg body weight, from 3.5 g/kg body weight to 5 g/kg body weight, from 4 glkg body weight to 5 g/kg body weight, or from 4.5 g/kg body weight to 5 g/kg body weight.
Effective doses may be extrapolated from dose-response curves derived from in vitro or animal model test bioassays or systems. The dosage should not be so large as to cause unacceptable adverse side effects.
[001051 In some embodiments, the dosage of a composition comprising a 12/15-LOX
inhibitor disclosed herein can be administered in a dose of from. about 20 mg/m2 to about 5,000 mg,m2 body surface area. For example, the dose can be from about 20 mg/m2 to about 200 mg/m2 body surface area; the dose can be from about 150 mg/m2 to about 500 mg/m2 body surface area; the dose can be from about 400 mg/m2 to about 1000 mg/m2 body surface area; the dose can be from about 900 mg/m2 to about 5,000 mg/m2 body surface area; the dose can be from about 200 mg/m2 to about 1,000 mg/m2 body surface area; or the dose can be from about 500 mg/m2 to about 600 mg/m2 body surface area.
Treatment [001061 Another aspect of the invention relates to the use of the compounds or compositions disclosed herein for the treatment of a condition involving 12/15-LOX. As used herein, the term "condition(s) involving 12/15-LOX" refers to any condition having 12/15-LOX as a cause of the condition, or a condition that can be worsened by the activity of 12/15-LOX, or a condition the progression of which is linked to the activity of 12/15-LOX. A condition involving 12/15-LOX
can therefore benefit therapeutically by inhibiting 12/15-LOX.
[001071 A method is provided herein for treating a condition involving 12/15-lipoxygenase in a subject, the method comprising administering to the subject an effective amount of a 12/15-LOX inhibitor disclosed herein. In methods of treatment described herein, the administration of 12/15-LOX inhibitor can be for either "prophylactic" or "therapeutic" purpose.
When provided prophylactically, the 12/15-LOX inhibitor is provided in advance of any symptom. When provided therapeutically, the 12/15-LOX inhibitor is provided at (or after) the onset of a symptom or indication of a condition involving 12/15-LOX. For example, stroke symptoms can include, but are not limited to, sudden numbness, tingling, weakness, or loss of movement in the face, arm, or leg, especially on only one side of the body; sudden vision changes; sudden trouble speaking; sudden confusion or trouble understanding simple statements; sudden problems with walking or balance; a sudden, severe headache that is different from past headaches.
[001081 12/15-LOX is an enzyme that is involved in several important diseases and conditions such as strokes, atherosclerosis, diabetes, obesity, asthma, glomerulonephritis, osteoporosis, and Alzheimer's. 12/15-LOX has recently been implicated in the disease of the newborn periventricular leukomal.acia, as well as in breast cancer growth. 12/15-LOX
has also been found.
to increase in mouse models of cardiac arrest with resuscitation, and mice in which the gene encoding 12/1.5-LOX has been deleted, are protected against brain injury in these mouse models.
Examples of conditions involving 12/15-LOX also include, but are not limited to, diseases involving apoptosis in cancer cells such as prostatic cancer, gastric cancer, breast cancer, pancreatic cancer, colorectal or esophageal cancer and airways carcinoma;
diseases involving hypoxia, or anoxia such as atherosclerosis, myocardial infarction, cardiovascular disease, heart failure (including chronic and congestive heart failure), cerebral ischemia, retinal ischemia, myocardial ischemia, post-surgical cognitive dysfunction and other ischemias;
diseases involving inflammation, including diabetes, arteriai inflammation, inflammatory bowel disease, Crohn's disease, renal disease, pre-menstrual syndrome, asthma, allergic rhinitis, gout; cardiopulmonary inflammation, rheumatoid arthritis, osteoarthritis, muscle fatigue and inflammatory disorders of the skin including acne, dermatitis and psoriasis; disorders of the airways such as asthma, chronic bronchitis, human airway carcinomas, mucus hypersecretion, chronic obstructive pulmonary disease (COPD), pulmonary fibrosis caused by chemotherapy or other drugs, idiopathic pulmonary fibrosis, cystic fibrosis, and adult respiratory distress syndrome; diseases involving central nervous system (CNS) disorders including psychiatric disorders including anxiety and depression; neurodegeneration and neuroinflammation including Alzheimer's, dementia and Parkinson's disease; peripheral neuropathy including spinal chord injury, head injury and surgical trauma, and allograft tissue and organ transplant rejection; diseases involving the autoitnmune system such as psoriasis, eczema, rheumatoid arthritis, and diabetes; and disorders involving bone loss or bone formation.
[001091 A suitable 12/15-LOX inhibitor can have an IC50 of less than 50 tiM, e.g., a suitable 12/15-LOX inhibitor can have an IC50 of from about 50 RM to about 5 nM, or less than 5 nM.

For example, in some embodiments, a suitable 12/15-LOX inhibitor has an IC50 of from about 50 ttM to about 25 p.M. from about 25 uM to about 10 uM, from about 10 uM to about 5 tiM, from about 5 ILM to about 11.1M, from about 1 ti.M to about 500 nM, from about 500 nM to about 400 nM, from about 400 nM to about 300 nM, from about 300 nM to about 250 nM, from about 250 nM to about 200 nM, from about 200 nM to about 150 nM, from about 150 nM
to about 100 nM, from about 100 nM to about 50 nM, from about 50 nM to about 30 nM, from about 30 nM
to about 25 nM, from about 25 nM to about 20 nM, from about 20 nM to about 15 nM, from about 15 nM to about 10 nM, from about 10 nM to about 5 nM, or less than about 5 nM.
[001101 The 12/15-LOX inhibitors of the present invention can also be used in applications known to benefit from the inhibition of 12/15-LOX. Non-limiting examples of these applications are disclosed in, US20060193797, US20120053220, US20060106014, US20060079488, US7576094, US20130131140, US2005070589, US20080207588, US2005065198, and US8048900.
[001111 With respect to duration and frequency of treatment, it is typical for skilled clinicians to monitor subjects in order to determine when the treatment is providing therapeutic benefit, and to determine whether to increase or decrease dosage, increase or decrease administration frequency, discontinue treatment, resume treatment or make other alteration to treatment regimen. The dosing schedule can vary from once a week to daily depending on a number of clinical factors, such as the subject's sensitivity to the drug. The desired dose can be administered at one time or divided into subdoses, e.g., 2-4 subdoses and administered over a period of time, e.g., at appropriate intervals through the day or other appropriate schedule. Such sub-doses can be administered as unit dosage forms. In some embodiments, administration is chronic, e.g., one or more doses daily over a period of weeks or months.
Examples of dosing schedules are administration daily, twice daily, three times daily or four or more times daily over a period of 1 week, 2 weeks, 3 weeks, 4 weeks, 1 month, 2 months, 3 months, 4 months, 5 months, or 6 months or more.
1001121 in some embodiments, the method is provided herein for treating a stroke in a subject using the 12/15-LOX inhibitors disclosed herein. For example, FIG. 5B shows that ML351 significantly reduces infarct size in a mouse model of permanent focal ischemia. Strokes are sudden neurological disorders that occur when blood flow to the brain is disturbed. There are two kinds of strokes. One is called acute ischemic stroke (AIS), which is due to blood flow blockage, Date Recue/Date Received 2020-11-17 and it accounts for 87% of strokes. A1S is an episode of neurological dysfunction caused by focal brain, spinal cord, or retina ischemia with evidence of acute infarction (Easton et al., Stroke 2009, 40, 2276-2293). There are at least four different causes of blood flow interruption: (1) a blood clot in a blood vessel; (2) a blood clot in the dural venous sinuses, which drain blood from the brain; (3) an embolus clogging a blood vessel; or (4) a sudden drop in blood pressure. Stroke symptoms can and frequently do persist beyond 24 hours if the patient survives the initial damage. The other kind of strokes is called hemorrhagic stroke, which is caused by a weakened blood vessel that ruptures and bleeds into the surrounding brain tissue. A
transient ischemic attack (T1A) is a condition mimicking AIS, in which a temporary interruption in blood flow to part of the brain results in impaired brain functions, but does not necessarily result in brain tissue damage.
[001131 Without wishing to be bound by theory, because oxidative stress is a major neurodegenerative process in ischemic diseases such as stroke, a role for 12/15-LOX in stroke-induced brain injury seemed reasonable and indeed, was found. Later work showed that 12/15-LOX was detrimental not just to neurons, but also to the brain vasculature after stroke, via a mechanism that involves an intracellular attack on mitochondria and translocation of the apoptosis-inducing factor (AlF) to the nucleus. Recent studies have shown that 12/15-LOX is also increased in human stroke (Yigitkanli et al., Ann. Neurol. 2013, 73, 129-135). In line with these findings, several LOX inhibitors have been found to reduce infarct size, leakage of the blood brain barrier and edema formation, and even hemorrhagic transformation following infusion of tPA.
1001141 Diagnostic methods for strokes include, but are not limited to, neuroimaging (e.g., magnetic resonance imaging (MR1), computerized tomography (CT), diffuse optical imaging, event-related optical signal, functional MR1, magnetoencephalography, positron emission tomography, or single-photon emission computed tomography), medical history, and physical exam.
1001151 In some embodiments, the 12/15-LOX inhibitors disclosed herein can be used in combination with other therapies to treat a stroke. An ischemic stroke can be treated by administering an antithrombotic agent to dissolve the blood clot.
Antithrombotic agents are further divided into the following three subtypes: anticoagulants, antiplatelet drugs, and thrombolytic drugs. Non-limiting examples of anticoagulants include:
cournarins, heparin, warfarin, acenocottmarol, phenprocoumon, atromentin, phenindione, fondaparinux, idraparinux, direct factor Xa inhibitors, direct thrombin inhibitors, antithrombin protein therapeutics, batroxobin, and hementin. Non-limiting examples of antiplatelet drugs include:
irreversible cyclooxygenase inhibitors (e.g., aspirin or triflusal), adenosine diphosphate receptor inhibitors (e.g., clopidogrel, prasugrel, ticagrelor, or ticlopidine), phosphodiesterase inhibitors (e.g., cilostazol), glycoprotein IIB/IHA inhibitors (e.g., abciximab, eptifibatide, or tirofiban), adenosine reuptake inhibitors (e.g., dipyridamole), and thrornboxane inhibitors (e.g., thromboxane synthase inhibitors or thromboxane receptor antagonists). Non-limiting examples of thrombolytic drugs include: tissue plasminogen activator t-PA. (e.g., alteplase, reteplase, or tenecteplase), anistreplase, streptokinase, and urokinase. An ischemic stroke can also be treated by endovascular procedures, in which a catheter is sent to the blood flow blockage site to remove the blood clot. t-PA can be optionally administered during the endovascular procedures.
[001161 In some embodiments, the 12/15-LOX inhibitors disclosed herein can be used to treat a stroke with hemorrhagic transformation. Hemorrhagic transformation refers to hemorrhages that develop inside areas of ischemia. For example, a subject who is on an oral anticoagulant and experience a stroke, can be treated with the 12/15-LOX inhibitors disclosed herein. In some embodiments, the 12/15-LOX inhibitors disclosed herein can be administered as adjuvant to tPA.
[001171 For the treatment of strokes, the 12/15-LOX inhibitors disclosed herein can be administered during the early phase of stroke or days (e.g., 1, 2, 3, 4, 5, 6,
7, 8, 9, 10, or more) after a stroke to aid in stroke recovery.
1001181 In some embodiments, the method is provided herein for treating periventricular leukomalacia (PVL) in a subject using the 12/15-LOX inhibitors disclosed herein. PVL patients also feature increased 12/15-LOX. Periventrieular leukomalacia is the most frequent cause of cerebral palsy in premature infants. This early neonatal disorder is due to the formation of single or multiple lesions of the ring of periventricular white matter, occurring during prenatal or neonatal life. Periventricular leukomalacia is responsible for the majority of motor sequelae of prematurity.
[001191 Another aspect of the invention relates to the use of the compounds described herein for the preparation of a medicament for the treatment of a condition involving 12/15-LOX.
[001201 It should be understood that this invention is not limited to the particular methodology, protocols, and reagents, etc., disclosed herein and as such may vary. The terminology used herein is for the purpose of describing particular embodiments only, and is not intended to limit the scope of the present invention, which is defined solely by the claims.
[001211 As used herein and in the claims, the singular forms include the plural reference and vice versa unless the context clearly indicates otherwise. Other than in the operating examples, or where otherwise indicated, all numbers expressing quantities of ingredients or reaction conditions used herein should be understood as modified in all instances by the term "about."
[001221 Although any known methods, devices, and materials may be used in the practice or testing of the invention, the methods, devices, and materials in this regard are disclosed herein.
[001231 Some embodiments of the invention are listed in the following paragraphs:
paragraph I A method of treating a condition involving 12/15-lipoxygenase in a subject, the method comprising administering to the subject an effective amount of a compound of Formula 1:

X
R11 ________________________ Formula I
or a pharmaceutically acceptable salt thereof, wherein: X is 0 or S; is an aryl, heteroaryl, cyclyl, or heterocyclyl, each of which can be optionally substituted; and R12 and R13 are independently hydrogen, halogen, alkyl, alkenyl, alkynyl, aralkyl, acyl, aryl, heteroaryl, cyclyl or heterocyelyl, each of which can be optionally substituted.
paragraph 2. The method of paragraph 1, wherein R11 is an aryl or heteroaryl, each of which can be optionally substituted.
paragraph 3. The method of paragraph 2, wherein RH is I-naphthyl, 2-naphthyl, isoquinolinyl, 2,3-dichlorophenyl, or 3,4-dichlorophenyl.
paragraph 4. The method of any of paragraphs 1-3, wherein R12 is hydrogen, alkyl, aralkyl, acyl, aryl, or heterocyclyl, each of which can be optionally substituted.
paragraph 5. The method of paragraph 4, wherein R12 is hydrogen, methyl, ethyl, propyl, butyl, pentyl, or phenyl.

paragraph 6. The method of any of paragraphs 1-5, wherein R13 is hydrogen, alkyl, aralkyl, acyl, aryl, or heterocyclyl, each of which can be optionally substituted.
paragraph 7. The method of paragraph 6, wherein R13 is hydrogen, methyl, ethyl, propyl, butyl, pentyl, or phenyl.
paragraph 8. The method of any of paragraphs 1-7, wherein R12 is hydrogen and R13 is hydrogen, methyl, ethyl, propyl, butyl, pentyl, or phenyl.
paragraph 9. The method of any of paragraphs 1-8, wherein X is 0.
paragraph 10. The method of any of paragraphs 1-8, wherein X is S.
paragraph 11. The method of paragraph 1, wherein the compound of Formula I is of:
(i) Formula II

R11¨c7 NH
\\N-4 CN
Formula II
, or a pharmaceutically acceptable salt thereof, wherein: X is 0 or S; and R11 is an aryl, heteroaryl, cyclyl, or heterocyclyl, each of which can be optionally substituted; or (ii) Formula III

\ /191 Formula III
, or a pharmaceutically acceptable salt thereof, wherein:
R12 and R13 are independently hydrogen, halogen, alkyl, alkenyl, alkynyl, aralkyl, acyl, aryl, heteroaryl, cyclyl or heterocyclyl, each of which can be optionally substituted.
paragraph 12. The method of paragraph 1, wherein the compound is selected from the group consisting of 5-(methylamino)-2-naphthalen-1-y1-1,3-oxazole-4-carbonitrile (1141351), 2-(2,3-dichloropheny1)-5-(methylamino)-1,3-oxazole-4-carbonitrile, 2-(3,4-dichloropheny1)-5-(methylamino)-1,3-oxazole-4-carbonitrile, 5-(methylamino)-2-naphthalen-l-y1-1,3-thiazole-4-carbonitrile.
paragraph 13. The method of paragraph 12, wherein the compound is 5-(methylamino)-2-naphthalen- I -y1-1,3-oxazole-4-carbonitrile (ML351).
paragraph 14. The method of any of paragraphs 1-13, wherein the compound inhibits 12/15-lipoxygenase.
paragraph 15. The method of any of paragraphs 1-14, wherein the condition is stroke, periventricular leukomalacia, cardiac arrest with resuscitation, atherosclerosis, Parkinson's disease, Alzheimer's disease, or breast cancer.
paragraph 16. The method of any of paragraphs 1-15, wherein the subject is a mammal.
paragraph 17. The method of any of paragraphs 1-16, wherein the subject is a human.
paragraph 18. Use of a compound of Formula I:
Ri2 R11 __ <
CN
Formula I
or a pharmaceutically acceptable salt thereof for the preparation of a medicament for the treatment of a condition involving 12/15-lipoxygenase, wherein:
X is 0 or S;
R11 is an aryl, heteroaryl, cyclyl, or heterocyclyl, each of which can be optionally substituted;
and R12 and R13 are independently hydrogen, halogen, alkyl, alkenyl, alkynyl, aralkyl, acyl, aryl, heteroaryl, cyclyl or heterocyclyl, each of which can be optionally substituted.
paragraph 19. The use paragraph 18, wherein the compound of Formula I is of:
(i) Formula II:

X
R11 __ ( NH
N-CN
Formula II
or a pharmaceutically acceptable salt thereof, wherein:
X is 0 or S; and Ri 1 is an aryl, heteroaryl, cyclyl, or heterocyclyl, each of which can be optionally substituted; or (ii) Formula III:

NCN
Formula III
or a pharmaceutically acceptable salt thereof, wherein:
RI2 and RI3 are independently hydrogen, halogen, alkyl, alkenyl, alkynyl, aralkyl, acyl, aryl, heteroaryl, cyclyl or heterocyclyl, each of which can be optionally substituted.
paragraph 20. The use of paragraph 18 or 19, wherein the compound is selected from the group consisting of 5-(methylamino)-2-naphthalen-l-y1-1,3-oxazole-4-carbonitrile (MI,351), dichloropheny1)-5-(methylamino)-1,3-oxazole-4-carbonitrile, 2-(3,4-dichloropheny1)-5-(methylamino)-1,3-oxazole-4-carbonitrile, and 5-(methylamino)-2-naphthalen-l-y1-1,3-thiazole-4-carbonitrile.
paragraph 21. The use of paragraph 18, wherein the condition is stroke, periventricular leukomalacia, cardiac arrest with resuscitation, atherosclerosis, Parkinson's disease, Alzheimer's disease, or breast cancer.
Definitions [001241 Unless stated otherwise, or implicit from context, the following terms and phrases include the meanings provided below. Unless explicitly stated otherwise, or apparent from context, the terms and phrases below do not exclude the meaning that the term or phrase has acquired in the art to which it pertains. The definitions are provided to aid in describing particular embodiments, and are not intended to limit the claimed invention, because the scope of the invention is limited only by the claims. Further, unless otherwise required by context, singular terms shall include pluralities and plural terms shall include the singular.
[001251 As used herein the term "comprising" or "comprises" is used in reference to compositions, methods, and respective component(s) thereof, that are useful to an embodiment, yet open to the inclusion of unspecified elements, whether useful or not.
[001261 As used herein the term "consisting essentially of" refers to those elements required for a given embodiment. The term permits the presence of elements that do not materially affect Eke basic and novel or functional characteristic(s) of that embodiment of the invention.
[001271 The terms "decrease", "reduce", or "inhibit" are all used herein to mean a decrease by a statistically significant amount. In some embodiments, "decrease", "reduce", or "inhibit"
typically means a decrease by at least 10% as compared to a reference level (e.g., the absence of a given treatment) and can include., for example, a decrease by at least about 10%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, at least about 99%, or more. A 12/15-LOX inhibitor can therefore decrease the biological function or activity of 12/15-LOX by at least about 10%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, at least about 99% , or more.
[00128.1 The term "disease", "disorder", or "condition" is used interchangeably herein, refers to any alternation in state of the body or of some of the organs, interrupting or disturbing the performance of the functions and/or causing symptoms such as discomfort, dysfunction, distress, or even death to the person afflicted or those in contact with a person. A
disease, disorder, or condition can also related to a distemper, ailing, ailment, malady, disorder, sickness, illness, complaint, affectation.
[001291 As used here, the term "pharmaceutically acceptable" refers to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
[001301 As used here, the term. "pharmaceutically-acceptable carrier" means a pharmaceutically-acceptable material, composition or vehicle, such as a liquid or solid filler, diluent, excipient, manufacturing aid (e.g., lubricant, talc magnesium., calcium or zinc stearate, or steric acid), or solvent encapsulating material, involved in carrying or transporting the subject compound from one organ, or portion of the body, to another organ, or portion of the body. Each carrier must be "acceptable" in the sense of being compatible with the other ingredients of the formulation and not injurious to the patient.
[001311 The phrase "therapeutically-effective amount" as used herein means that amount of a compound, material, or composition which is effective for producing some desired therapeutic effect in at least a sub-population of cells in an animal at a reasonable benefit/risk ratio applicable to any medical treatment. For example, an amount of a compound administered to a subject that is sufficient to produce a statistically significant, measurable change in at least one symptom of strokes. For example, the brain infarct size is decreased by at least about 5%, at least about 10A, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, at least about 99% , or more.
Determination of a therapeutically effective amount is well within the capability of those skilled in the art. Generally, a therapeutically effective amount can vary with the subject's history, age, condition, sex, as well as the severity and type of the medical condition in the subject, and administration of other pharmaceutically active agents.
[001321 As used herein, the term "administer" refers to the placement of a inhibitor into a subject by a method or route which results in at least partial localization of the 12/15-LOX inhibitor at a desired site such that desired effect is produced. A
compound or composition described herein can be administered by any appropriate route known in the art including, but not limited to, oral or parenteral routes, including intravenous, intramuscular, subcutaneous, transdermal, airway (aerosol), pulmonary, nasal, rectal, and topical (including buccal and sublingual) administration.
[001331 Exemplay modes of administration include, but are not limited to, injection, infusion, instillation, inhalation, or ingestion. "Injection" includes, without limitation, intravenous, intramuscular, intraarterial, intrathecal, intraventricular, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, sub capsular, subarachnoid, intraspinal., intracerebro spinal, and intrastemal injection and infusion.
In preferred embodiments, the compositions are administered by intravenous infusion or injection.
1001341 By "treatment", "prevention" or "amelioration" of a disease, disorder, or condition is meant delaying or preventing the onset of such a disease or disorder, reversing, alleviating, ameliorating, inhibiting, slowing down or stopping the progression, aggravation or deterioration the progression or severity of a condition associated with such a disease, disorder, or condition.
The term "treatment" of a disease, disorder, or condition also includes providing relief from the symptoms or side-effects of the disease (including palliative treatment. In one embodiment, the symptoms of a disease, disorder, or condition are alleviated by at least 5%, at least 10%, at least 20%, at least 30%, at least 40%, or at least 50%.
1001351 As used herein, a "subject" means a human or animal. Usually the animal is a vertebrate such as a primate, rodent, domestic animal or game animal. Primates include chimpanzees, cynomologous monkeys, spider monkeys, and macaques, e.g., Rhesus.
Rodents include mice, rats, woodchucks, ferrets, rabbits and hamsters. Domestic and game animals include cows, horses, pigs, deer, bison, buffalo, feline specks, e.g., domestic cat, and canine species, e.g., dog, fox, wolf. The terms, "patient", "individual" and "subject" are used interchangeably herein.
1001361 Preferably, the subject is a mammal. The mammal can be a human, non-human primate, mouse, rat, dog, cat, horse, or cow, but arc not limited to these examples.
1001371 Certain compounds of the present invention and definitions of specific functional groups are also described in more detail below. For purposes of this invention, the chemical elements are identified in accordance with the Periodic Table of the Elements, CAS version, Handbook of Chemistry and Physics, 75th Ed., inside cover, and specific functional groups are generally defined as described therein. Additionally, general principles of organic chemistry, as well as specific functional moieties and reactivity, are described in Organic Chemistry, Thomas Sorrell, University Science Books, Sausalito: 1999.
[00138] As used herein, the term "aliphatic" means a moiety characterized by a straight or branched chain arrangement of constituent carbon atoms and can be saturated or partially unsaturated with one or more (e.g., one, two, three, four, five or more) double or triple bonds.
[00139] As used herein, the term "alicyclic" means a moiety comprising a nonaromatic ring structure. Alicyclic moieties can be saturated or partially unsaturated with one or more double or triple bonds. Alicyclic moieties can also optionally comprise heteroatoms such as nitrogen, oxygen and sulfur. The nitrogen atoms can be optionally quaternerized or oxidized and the sulfur atoms can be optionally oxidized. Examples of alicyclic moieties include, but are not limited to moieties with C3-C8 rings such as cyclopropyl, cyclohexane, cyclopentane, cyclopentene, cyclopentadiene, cyclohexane, cyclobexene, cyclohexadiene, cycloheptane, cycloheptene, cycloheptadiene, cyclooctane, cyclooctene, and cyclooctadiene.
[00140] The term, "heteroatom" refers to an atom that is not a carbon atom.
Particular examples of heteroatoms include, but are not limited to nitrogen, oxygen, sulfur and halogens. A
"heteroatom moiety" includes a moiety where the atom by which the moiety is attached is not a carbon. Examples of heteroatom moieties include ¨N=, ¨NRN¨, ¨N+(0)=, ¨0¨, ¨S¨
or ¨S(0)2¨, ¨0S(0)2¨, and ¨SS¨, wherein RN is H or a further substituent.
[00141] As used herein, the term "alkyl" means a straight or branched, saturated aliphatic radical having a chain of carbon atoms. Cx alkyl and C-C, alkyl are typically used where X and Y indicate the number of carbon atoms in the chain. For example, C1-C6 alkyl includes alkyls that have a chain of between 1 and 6 carbons (e.g., methyl, ethyl, propyl, isopropyl, butyl, sec-butyl, isobutyl, tert-butyl, pentyl, neopentyl, hexyl, and the like). Alkyl represented along with another radical (e.g., as in arylalky, I) means a straight or branched, saturated alkyl divalent radical having the number of atoms indicated or when no atoms are indicated means a bond, e.g., (C6-C10)aryl(Co-C3)alkyl includes phenyl, benzyl, phenethyl, 1-phenylethyl 3-phenylpropyl, and the like. Backbone of the alkyl can be optionally inserted with one or more heteroatoms, such as N, 0, or S.

Date Recue/Date Received 2020-11-17 [001421 In preferred embodiments, a straight chain or branched chain alkyl has 30 or fewer carbon atoms in its backbone (e.g., Cl-C30 for straight chains, C3-C30 for branched chains), and more preferably 20 or fewer. Likewise, preferred cycloalkyls have from 3-10 carbon atoms in their ring structure, and more preferably have 5, 6 or 7 carbons in the ring structure. The term "al.kyl" (or "lower alkyl") as used throughout the specification, examples, and claims is intended to include both "unsubstituted alkyls" and "substituted alkyls", the latter of which refers to alkyl moieties having one or more substituents replacing a hydrogen on one or more carbons of the hydrocarbon backbone. In some embodiments, a straight chain or branched chain alkyl has 5 or fewer carbon atoms, 10 or fewer carbon atoms, or 15 or fewer carbon atoms.
[001431 Unless the number of carbons is otherwise specified, "lower alkyl" as used herein means an alkyl group, as defined above, but having from one to ten carbons, more preferably from. one to six carbon atoms in its backbone structure. Likewise, "lower alkenyl" and "lower alkynyl" have similar chain lengths. Throughout the application, preferred alkyl groups are lower alkyls. In preferred embodiments, a substituent designated herein as alkyl is a lower alkyl.
[001441 Substituents of a substituted alkyl can include halogen., hydroxy, nitro, thiols, amino, azido, imino, amido, phosphoryl. (including phosphonate and phosphinate), sulfonyl. (including sulfate, sulfonamido, sulfamoyl and sulfonate), and silyl groups, as well as ethers, alk.ylthios, carbonyls (including ketones, aldehydes, carboxyl.ates, andesters),-CF3, -CN
and the like. In some embodiments, the substituted alkyl is a perfluorinated alkyl.
[001451 As used herein, the term "alkenyl" refers to unsaturated straight-chain, branched-chain or cyclic hydrocarbon radicals having at least one carbon-carbon double bond. Cx alkenyl and C1-Cy alkenyl are typically used where X and Y indicate the number of carbon atoms in the chain. For example, C2-C6a1keny1 includes alkenyls that have a chain of between 1 and 6 carbons and at least one double bond, e.g., vinyl, ally!, propenyl, isopropenyl, 1-butenyl, 2-butenyl, 3-butenyl, 2-methylallyl, 1-hexenyl, 2-hexenyl, 3- hexenyl, and the like).
Alkenyl represented along with another radical (e.g., as in arylalkenyl) means a straight or branched, alkenyl divalent radical having the number of atoms indicated. Backbone of the alkenyl can be optionally inserted with one or more heteroatoms, such as N, 0, or S. In some embodiments, the substituted alkenyl is a perfluorinated alkenyl.
[001461 As used herein, the term "alkynyl" refers to unsaturated hydrocarbon radicals having at least one carbon-carbon triple bond. C, alkynyl and C3-Cy alkynyl are typically used where X

and Y indicate the number of carbon atoms in the chain. For example, C2-Coa1kyny1 includes alkynis that have a chain of between 1 and 6 carbons and at least one triple bond, e.g., ethynyl, 1-propynyl, 2-propynyl, 1-butynyl, isopentynyl, 1,3-hexa-diyn-yl, n-hexynyl, 3-pentynyl, 1-hexen-3-ynyl and the like. Alkynyl represented along with another radical (e.g., as in arylalkynyl) means a straight or branched, alkynyl divalent radical having the number of atoms indicated.
Backbone of the alkynyl can be optionally inserted with one or more heteroatoms, such as N, 0, or S. In some embodiments, the substituted alkynyl is a perfluorinated alkynyl.
[001471 The terms "alkylene," "alkenylene," and "alkynylene" refer to divalent alkyl, alkelyne, and alkynylene" radicals. Prefixes Cx and Cx-Cy are typically used where X and Y
indicate the number of carbon atoms in the chain. For example, C1-C6alkylerie includes methylene, (¨CH2¨), ethylene (¨CH2CH2¨), trimethylene (¨CH2CH2CH2--), tetramethylene (---CH2CH2CH2CH2¨), 2-methyltetramethylene ( -CH2CH(CH3)CH2CHL
pentamethylene (--CH2CII2CH2CH2CH2¨) and the like).
[001481 As used herein, the term "alkylidene" means a straight or branched unsaturated, aliphatic, divalent radical having a general formula =C12.14. C alkylidene and Cx-Cyalkylidene are typically used where X and Y indicate the number of carbon atoms in the chain. For example, C2-C6alkylidene includes methylidene (=CH2), ethylidene (=CHCH3), isopropylidene (=C(CH3)2), propylidene (=CHCH2CH3), allylidene (=CH CH=CH2), and the like).
[001491 As used herein, the term "halogen" or "halo" refers to an atom selected from fluorine, chlorine, bromine and iodine.
1001501 The term "aryl" refers to monocyclic, bicyclic, or tricyclic fused aromatic ring system. Cõ aryl and C,-Cy aryl are typically used where X and Y indicate the number of carbon atoms in the ring system. An aryl group can comprise a 4-atom ring, a 5-atom ring, a 6-atom ring, a 7-atom ring, a 8-atom ring, a 9 atom ring, or more. Exemplary aryl groups include, but are not limited to, pyridinyl, pyrimidinyl, furanyl, thienyl, imidazolyi, thiazolyl, pyrazolyl, pyridazinyl, pyrazinyl, triazinyl, tetrazolyl, indolyl, benzyl, phenyl, naphthyl, anthracenyl, azulenyl, fluorenyl, indanyl, indenyl, naphthyl, phenyl, tetrahydronaphthyl, benzimidazolyl, benzofuranyl, benzothiofuranyl, benzothiophenyl, benzoxazolyl, benzoxazolinyl, benzthiazolyl, benztriazolyl, benztetrazolyl, benzisoxazolyl, benzisothiazolyl, benzimidazolinyl, carbazolyl, 4aH carbazolyl, carbolinyl, chromanyl, chromenyl, cinnolinyl, decahydroquinolinyl, 2H,6H-1,5,2-dithiazinyl, dihydrofuro[2,3 b]tetrahydrofuran, fitranyl, fitrazanyl, imidazolidinyl, imidazolinyl, imidazolyl, 1H-indazolyl, indolenyl, indolinyl, indolizinyl, indolyl, 3H-indolyl, isatinoyl, isobenzofuranyl, isochmmanyl, isoindazolyl, isoindolinyl, isoindolyl, isoquinolinyl, isothiazolyl, isoxazolyl, methylenedioxyphenyl, morpholinyl, naphthyridinyl, octahydroisoquinolinyl, oxadiazolyl, 1,2,3-oxadiazoly1,1,2,4-oxadiazolyl, 1,2,5-oxadiazolyl, 1,3,4-oxadiazolyl, oxazolidinyl, oxazolyl, oxindolyl, pyTimidinyl, phenanthridinyl, phenanthrolinyl, phenazinyl, phenothiazinyl, phenoxathinyl, phenoxazinyl., phthalazinyl, piperazinyl., piperidinyl, piperidonyl, 4-piperidonyl, piperonyl, pteridinyl, purinyl, pyranyl., pyrazinyl., pyrazolidinyl, pyrazolinyl, pyrazolyl, pyridazinyl, pyridooxazole, pyridoimidazole, pyridothiazole, pyridinyl, pyridyl, pyrimidinyl, pyrrolidinyl, pyrrolinyl, 2H-pyrrolyi, pyrrolyl, quinazolinyl, quinolinyl, 4H-quinolizinyl, quinoxalinyl., quinuclidinyl, tetrahydrofuranyl, tetrahydroisoquinolinyl, tetrahydroquinolinyl, tetrazol.yl, 6H-1,2,5-thiadiazinyl., thiadiazolyl, 1,2,4-thiadiazol.yl, 1,2,5-thiadiazolyl, 1,3,4-thiadiazolyl, thianthrenyl, thiazolyl, thienyl, thienothiazolyl, thienooxazolyl, thienoimidazolyl, thiophenyl and xanthenyl, and the like. In some embodiments, 1, 2, 3, or 4 hydrogen atoms of each ring can be substituted by a substituent.
1001511 The term "heteroaryl" refers to an aromatic 5-8 membered monocyclic, 8-membered fused bicyclic, or 11-14 membered fused tricyclic ring system having 1-3 heteroatoms if monocyclic, 1-6 heteroatoms if bicyclic, or 1-9 heteroatoms if tricyclic, said heteroatoms selected from 0, N, or S (e.g., carbon atoms and 1-3, 1-6, or 1-9 heteroatoms of N, 0, or S if monocyclic, bicyclic, or tricyclic, respectively. Cx heteroaryl and C1-Cyheteroaryl are typically used where X and Y indicate the number of carbon atoms in the ring system.
Heteroaryls include, but are not limited to, those derived from benzo[b]furan, benzo[b]
thiophene, benzimidazole, imida70[4,5-c)pyridine, quinazoline, thieno(2,3-c)pyridine, thieno[3,2-bipyridine, thieno[2, 3-b]pyridine, indolizine, imidazo[1,2alpyridine, quinoline, isoquinoline, phthalazine, quinoxaline, naphthyridine, quinolizine, indole, isoindole, indazole, indoline, benzoxazole, benzopyrazole, benzothiazole, imidazo[1,5-a]pyridine, pyrazolo[I,5-a]pyridine, imidazo[1,2-a]pyrimidine, imidazo[1,2-dpyrimidine, imidazo[1,5-a]pyrimidine, imidazo[1,5-c]pyrimidine, pyrrolo[2,3-b]pyridine, pyrrolo[2,3cjpyridine, pyrrolo[3,2-c]pyridine, pyrrolo[3,2-b]pyridine, pyrrolo[2,3-cl]pyrimidine, pyrrolo[3,2-d]pyrimidine, pyrrolo [2,3-b]pyrazine, pyrazolo[1,5-a]pyridine, pyrrolo[1,2-b]pyridazine, pyrrolo[1,2-c]pyrimidine, pyrrolo[1,2-a]pyrirnidine, pyrrolo[1,2-a]pyrazine, triazo[1,5-a]pyridine, pteridine, purine, carbazole, acridine, phenazine, phenothiazene, phenoxazine,1,2-dihydropyrrolo[3,2,1-hi]indole, indolizine, pyrido[I,2-a]indole, 2(IH)-pyridinone, benzimidazolyl, benzofuranyl, benzothiofuranyl, benzothiophenyl, benzoxazolyl, benzoxazolinyl, benzthiazolyl, benztriazolyl, benztetrazolyl, benzisoxazolyl, benzisothiazolyl, benzimidazolinyl, carbazolyl, 4aH-carbazolyl, carbolinyl, chromanyl, chromenyl, cinnolinyl, decahydroquinolinyl, 2H,6H- I ,5,2-dithiazinyl, dihydrofuro[2,3-b]tetrahydrofuran, furanyl, furazanyl, imidazolidinyl, imidaz.olinyl, imidazolyl, IH-indazolyl, indolenyl, indolinyl, indolizinyl, indolyl, 3H-indolyl, isatinoyl, isobenzofuranyl, isochromanyl, isoindazolyl, isoindolinyl, isoindolyl, isoquinolinyl, isothiazolyl, isoxazolyl, methylenedioxyphenyl, morpholinyl, naphthyridinyl, octahydroisoquinolinyl, oxadiazolyl, 1,2,3-oxadiazolyl, 1,2,4-oxadiazolyl, 1,2,5-oxadiazolyl, 1,3,4-oxadiazolyl, oxazolidinyl, oxazolyl, oxepanyl, oxetanyl, oxindolyl, pyrimidinyl, phenanthridinyl, phenanthrolinyl, phenazinyl, phenothiazinyl, phenoxathinyl, phenoxazinyl, phthalazinyl, piperazinyl, piperidinyl, piperidonyl, 4-piperidonyl, piperonyl, pteridinyl, purinyl, pyranyl, pyrazinyl, pyraz,olidinyl, pyrazolinyl, pyrazolyl, pyridazinyl, pyridooxazole, pyridoimidazole, pyridothiazole, pyridinyl, pyridyl, pyrimidinyl, pyrrolidinyl, pyrrolinyl, 2H-pyrrolyl, pyrrolyl, quinazolinyi, quinolinyl, 41-1-quinolizinyl, quinoxalinyl, quinuclidinyl, tetrahydrofuranyl, tetrahydroisoquinolinyl, tetrahydropyranyl, tetrahydroquinolinyl, tetrazolyl, 6H-1,2,5-thiadiazinyl, 1,2,3-thiadiazolyl, I ,2,4-thiadiazolyl, 1,2,5-thiadiazolyl, 1,3,4-thiadiazolyi, thianthrenyl, thiazolyl, thienyl, thienothiazolyl, thienooxazolyl, thienoimidazolyl, thiophenyl and xanthenyl.
Some exemplary heteroaryl groups include, but are not limited to, pyridyl, furyl or furanyl, imidazolyl, benzimidazolyl, pyrimidinyl, thiophenyl or thienyl, pyridazinyl, pyrazinyl, quinolinyl, indolyl, thiazolyl, naphthyridinyl, 2-amino-4-oxo-3,4-dihydropteridin-6-yl, tetrahydroisoquinolinyl, and the like. In some embodiments, 1, 2, 3, or 4 hydrogen atoms of each ring may be substituted by a substituent 1001521 Aryl and heteroaryls can be optionally substituted with one or more substituents at one or more positions, for example, halogen, alkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, hydroxyl, amino, nitro, sulthydryl, imino, amido, phosphate, phosphonate, phosphinate, carbonyl, carboxyl, silyl, ether, alkylthio, sulfonyl, ketone, aldehyde, ester, a heterocyclyl, an aromatic or heteroaromatic moiety, -CF3, -CN, or the like.
[001531 The term "cycly1" or "cycloalkyl" refers to saturated and partially unsaturated cyclic hydrocarbon groups having 3 to 12 carbons, for example, 3 to 8 carbons, and, for example, 3 to 6 carbons. C.cycly1 and C.-Cycylcyl are typically used where X and Y indicate the number of carbon atoms in the ring system. The cycloalkyl group additionally can be optionally substituted, e.g., with 1, 2, 3, or 4 substituents. C3-C1ocycly1 includes cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclohexenyl, 2,5-cyclohexadienyl, cycloheptyl, cyclooctyl, bicyclo[2.2.2]octyl, adamantan-l-yl, decahydronaphthyl, oxocyclohexyl, dioxocyclohexyl, thiocyclohexyl, 2-oxobicyclo [2.2.1]hept-l-yl, and the like.
[001541 The term "heterocyclyl" refers to a nonaromatic 5-8 membered monocyclic, 8-12 membered bicyclic, or 11-14 membered tricyclic ring system having 1-3 heteroatoms if monocyclic, 1-6 heteroatoms if bicyclic, or 1-9 heteroatoms if tricyclic, said heteroatoms selected from. 0, N, or S (e.g., carbon atoms and 1-3, 1-6, or 1-9 heteroatoms of N, 0, or S if monocyclic, bicyclic, or tricyclic, respectively). C. heterocyclyl and Cx-Cy heterocyclyl are typically used where X and Y indicate the number of carbon atoms in the ring system. In some embodiments, I, 2 or 3 hydrogen atoms of each ring can be substituted by a substituent.
Exemplary heterocyclyl groups include, but are not limited to piperazinyl, pyrrolidinyl, dioxanyl, morpholinyl, tetrahydrofuranyl, piperidyl, 4-morpholyl, 4-piperazinyl, pyrrolidinyl, perhydropyrrolizinyl, 1,4-diazaperhydroepinyl, 1,3-dioxanyl, 1,4-dioxanyland the like.
1001551 The terms "bicyclic" and "tricyclic" refers to fused, bridged, or joined by a single bond polycyclic ring assemblies.
1001561 The term "cyclylalkylene" means a divalent aryl, heteroaryl, cyclyl, or heterocyclyl.
1001571 As used herein, the term, "aromatic" means a moiety wherein the constituent atoms make up an unsaturated ring system, all atoms in the ring system are sp2 hybridized and the total number of pi electrons is equal to 4n+2. An aromatic ring canbe such that the ring atoms are only carbon atoms (e.g., aryl) or can include carbon and non-carbon atoms (e.g., heteroaryl).
[001581 As used herein, the term "substituted" refers to independent replacement of one or more (typically 1, 2, 3, 4, or 5) of the hydrogen atoms on the substituted moiety with substituents independently selected from the group of substituents listed below in the definition for "substituents" or otherwise specified. In general, a non-hydrogen substituent can be any substituent that can be bound to an atom of the given moiety that is specified to be substituted.
Examples of substituents include, but are not limited to, acyl, acylamino, acyloxy, aldehyde, alicyclic, aliphatic, alkanesulfonamido, alkariesulfonyl, alkaryl, alkenyl, alkoxy, alkoxycarbonyl, alkyl, alkylarnino, alkylcarbanoyl, alkylene, alkylidene, alkylthios, alkynyl, amide, amido, amino, arninoalkyl, aralkyl, aralkylsulfonamido, arenesulfonamido, arenesulfonyl, aromatic, aryl, arylamino, arylcarbanoyl, aryloxy, azido, earbamoyl, carbonyls (including ketones), carboxy, carboxylates, CF3, cyano (CN), cycloalkyl, cycloalkylene, ester, ether, haloalkyl, halogen, halogen, heteroaryl., heterocyclyl, hydroxy, hydroxy, hydroxyalkyl, imino, iminoketone, ketone, mercapto, nitro, oxaalkyl, oxo, oxoalkyl, phosphoryl (including phosphonate and phosphinate), silyl groups, sulfonamido, sulfonyl. (including sulfate, sulfam.oyl and sulfonatc), thiols, and ureido moieties, each of which may optionally also be substituted or unsubstituted. in some cases, two substituents, together with the carbon(s) to which they are attached to, can. form a ring.
R01591 The terms "alkoxyl" or "alkoxy" as used herein refers to an alkyl group, as defined above, having an oxygen radical attached thereto. Representative alkoxyl groups include methoxy, ethoxy, propyloxy, tert-butoxy, n-propyloxy, iso-propyloxy, n-butyloxy, iso-butyloxy, and the like. An "ether" is two hydrocarbons covalent"), linked by an. oxygen.
Accordingly, the substituent of an alkyl that renders that alkyl an ether is or resembles an alkoxyl., such as can be represented by one of -0-alkyl, -0-alkenyl, and -0-alkynyl. Aroxy can be represented by ¨0-aryl or 0-heteroaryl, wherein aryl and heteroaryl are as defined below. The alkoxy and aroxy groups can be substituted as described above for alkyl.
1001601 The term "aralkyl", as used herein, refers to an alkyl group substituted with an aryl group (e.g., an aromatic or heteroaromatic group).
1001611 The term "alkylthio" refers to an alkyl group, as defined above, having a sulfur radical attached thereto. in preferred embodiments, the "alkylthio" moiety is represented by one of -S-alkyl, -S-alkenyl, and -S-alkynyl. Representative alkylthio groups include methylthio, ethylthio, and the like. The term "alkylthio" also encompasses cycloalkyl groups, alkene and cycloalkene groups, and allcyne groups. "Arylthio" refers to aryl or heteroaryl groups.
1001621 As used herein, the term "amino" means -NH2. The term "alkylamino"
means a nitrogen moiety having at least one straight or branched unsaturated aliphatic, cyclyl, or heterocyclyl radicals attached to the nitrogen. For example, representative amino groups include ¨NH2, ¨NHCH3, ¨N(CH3)2, ¨NH(Ci-Cmalkyl), ¨N(Ci-Cloalky1)2, and the like. The term "alkylamino" includes "alkenylamino," "allcynylamino," "cyclylamino," and "heterocyclylamino." The term "arylamino" means a nitrogen moiety having at least one aryl radical attached to the nitrogen. For example ¨NHaryl, and ¨N(aryl)2. The term "heteroarylamino" means a nitrogen moiety having at least one heteroaryl radical attached to the nitrogen. For example ¨NHheteroaryl, and ¨N(heteroary1)2. Optionally, two substituents together with the nitrogen can also form a ring. Unless indicated otherwise, the compounds described herein containing amino moieties can include protected derivatives thereof. Suitable protecting groups for amino moieties include acetyl, tertbutoxycarbonyl, benzyloxycarbonyl, and the like.
[001631 The term. "aminoalkyl" means an alkyl, alkenyl, and alkynyl as defined above, except where one or more substituted or unsubstituted nitrogen atoms (¨N¨) are positioned between carbon atoms of the alkyl, alk.enyl, or alkynyl . For example, an (C2-C6) aminoalkyl refers to a chain comprising between 2 and 6 carbons and one or more nitrogen atoms positioned between the carbon atoms.
[001641 As used herein, the term "carbonyl" means the radical C(0)---. It is noted that the carbonyl radical can be further substituted with a variety of substituents to form different carbonyl groups including acids, acid halides, amides, esters, ketones, and the like.
[001651 The term "carboxyl" refers to a functional group with the formula -00011.
[001661 The term "carboxy" means the radical ---C,(0)0 . It is noted that compounds described herein containing carboxy moieties can include protected derivatives thereof, i.e., where the oxygen is substituted with a protecting group. Suitable protecting groups for carboxy moieties include benzyl, tert-butyl, and the like.
1001671 The term "hydroxyl" means the radical ¨OH.
[001681 The term "nitro" means the radical ¨NO2.
1001691 The term "sulfonyl" means the radical ¨SO2¨. It is noted that the sulfonyl radical can be further substituted with a variety of substituents to form different sulfonyl groups including sulfonic acids (-S03H), sulfonamides, sulfonate esters, sulfones, and the like.
[001701 As used herein, the term "pharmaceutically-acceptable salts" refers to the conventional nontoxic salts or quaternary ammonium salts of therapeutic agents, e.g., from non-toxic organic or inorganic acids. These salts can be prepared in situ in the administration vehicle or the dosage form manufacturing process, or by separately reacting a therapeutic agent in its free base or acid form with a suitable organic or inorganic acid or base, and isolating the salt thus formed during subsequent purification. Conventional nontoxic salts include those derived from inorganic acids such as sulfuric, sulfamic, phosphoric, nitric, and the like;
and the salts prepared from organic acids such as acetic, propionic, succinic, glycolic, stearic, lactic, malic, tartaric, citric, ascorbic, palmitic, maleic, hydroxymaleic, phenylacetic, glutamic, benzoic, salicyclic, sulfanilic, 2-acetoxybenzoic, fumaricõ toluenesulfonic, methanesulfonic, ethane disulfonic, oxalic, isothionic, and the like. See, for example, Berge et al., "Pharmaceutical Salts", J. Phann.
Sci. 66:1-19 (1977).
[001711 In some embodiments of the aspects described herein, representative salts include the hydrobromide, hydrochloride, sulfate, bisulfate, phosphate, nitrate, acetate, succinate, valerate, oleate, palmitate, stearate, laurate, benzoate, lactate, phosphate, tosylate, citrate, maleate, fumarate, succinate, tartrate, napthylate, mesylate, glucoheptonate, lactobionate, and laurylsulphonate salts and the like.
100172] The singular terms "a," "an," and "the" include plural referents unless context clearly indicates otherwise. Similarly, the word "or" is intended to include "and"
unless the context clearly indicates otherwise.
[00173] Other than in the operating examples, or where otherwise indicated, all numbers expressing quantities of ingredients or reaction conditions used herein should be understood as modified in all instances by the term "about." The term "about" when used in connection with percentages may mean 5% of the value being referred to. For example, about 100 means from 95 to 105.
(00174) Although methods and materials similar or equivalent to those disclosed herein can be used in the practice or testing of this disclosure, suitable methods and materials are described below. The term "comprises" means "includes." The abbreviation, "e.g." is derived from the Latin exempli gratia, and is used herein to indicate a non-limiting example.
Thus, the abbreviation "e.g." is synonymous with the term "for example."
1001751 Although preferred embodiments have been depicted and described in detail herein, it will be apparent to those skilled in the relevant art that various modifications, additions, substitutions, and the like can be made without departing from the spirit of the invention and these are therefore considered to be within the scope of the invention as defined in the claims which follow. Further, to the extent not already indicated, it will be understood by those of ordinary skill in the art that any one of the various embodiments herein described and illustrated can be further modified to incorporate features shown in any of the other embodiments disclosed herein.

Date Recue/Date Received 2020-11-17 [00176] All patents and other publications; including literature references, issued patents, published patent applications, and co-pending patent applications; cited throughout this application are expressly for the purpose of describing and disclosing, for example, the methodologies described in such publications that might be used in connection with the technology disclosed herein. These publications are provided solely for their disclosure prior to the filing date of the present application. Nothing in this regard should be construed as an admission that the inventors are not entitled to antedate such disclosure by virtue of prior invention or for any other reason. All statements as to the date or representation as to the contents of these documents is based on the information available to the applicants and does not constitute any admission as to the correctness of the dates or contents of these documents.
[001771 The description of embodiments of the disclosure is not intended to be exhaustive or to limit the disclosure to the precise form disclosed. While specific embodiments of, and examples for, the disclosure are disclosed herein for illustrative purposes, various equivalent modifications are possible within the scope of the disclosure, as those skilled in the relevant art will recognize. For example, while method steps or functions are presented in a given order, alternative embodiments may perform functions in a different order, or functions may be performed substantially concurrently. The teachings of the disclosure provided herein can be applied to other procedures or methods as appropriate. The various embodiments disclosed herein can be combined to provide further embodiments. Aspects of the disclosure can he modified, if necessary, to employ the compositions, functions and concepts of the above references and application to provide yet further embodiments of the disclosure.
10178) Specific elements of any of the foregoing embodiments can be combined or substituted for elements in other embodiments. Furthermore, while advantages associated with certain embodiments of the disclosure have been described in the context of these embodiments, other embodiments may also exhibit such advantages, and not all embodiments need necessarily exhibit such advantages to fall within the scope of the disclosure.
EXAMPLES
(00179) The following examples illustrate some embodiments and aspects of the invention. It will be apparent to those skilled in the relevant art that various modifications, additions, substitutions, and the like can be performed without altering the spirit or scope of the invention, Date Recue/Date Received 2020-11-17 and such modifications and variations are encompassed within the scope of the invention as defined in the claims which follow. The following examples do not in any way limit the invention.
1001801 The technology disclosed herein is further illustrated by the following examples which in no way should be construed as being further limiting.
Example 1: Potent and Selective Inhibitors of Human .Reticulocyte 12/15-Lipoxygenase [001811 A key challenge facing drug discovery today is variability of the drug target between species, such as with 12/15-lipoxygenase (12/15-LOX), which contributes to ischemic brain injury, but its human and rodent isozymes have different inhibitor specificities. Described herein, the inventors have utilized a quantitative high-throughput (qHTS) screen to identify, among other things, compound 1 (ML351), a novel chetnotype for 12/15-LOX inhibition, which has nanomolar potency (ICso = 200 nM) against human 12/15-LOX and is protective against oxidative glutamate toxicity in mouse neuronal HT-22 cells. In addition, it exhibited greater than 250-fold selectivity versus related LOX isozymes, was a mixed inhibitor, and did not reduce the active-site ferric ion. Finally, 1 significantly reduced infarct size following permanent focal ischemia in a mouse model of ischemic stroke. As such, this represents the first report of a selective inhibitor of human 12/15-LOX with demonstrated in vivo activity in proof-of-concept mouse models of stroke.
[001821 Chemistry 1001831 The compounds 1-18 from Table 1 were synthesized as shown in FIG. 1A.
The intermediates Ii-18i and 19 were obtained by propylphosphonic anhydride (T3I)(5 assisted coupling of the corresponding commercially available carboxylic acids with 2-aminomalanonitrile, followed by cyclization in one step. This modified synthesis of 5-amino-2-substituted-1,3-oxazole-4-carbonitrils allows facile access to a variety of compounds via a simple work up and purification from commercially available carboxylic acids.
Due to low nuckophilicity of the amine group, the initial attempts to monomethylate the 5-amino-2-substituted-1,3-oxazole-4-carbonitrile intermediate with several known methylation methods including methyl iodide, formic acid or methyl boronic acid were met with limited success.
However, condensation with paraformaldehyde in the presence of sodium methoxide in methanol, followed by in situ reduction with sodium borohydride, afforded the required products 1-18 in modest yields.
Table 1. Variations to compound 1 (analogs 2-18):
1 fYle sa. NH
1 N- =====.t.iN
IC:50 (gM) Compound R 1050 (iuM}
Compound R
ick SD ($1.14)1 s.s., __________________________________ 0õ -..k.õ.'=Ii.
[21 1 L....)%0J 0.2() '7.6 ¨
to,-..reryk-, 4.._(....) >40 2 i i it =>3(/' ii pe --Ph re, 1,-,.. =)k.N.:Ii.
23 [13j ...:.,-...:..- H
4 ,...4s-N.:.
,= : ,.., 3,6 [0.51 13 A :1 `...--k,...0 )40 ...ik=¨.) `-=' ?..-1: 0.73 14 io-'1--- 3.9 f 0.21 r:l...kõ..4.,,2 H
.H., ., 6 r ts >40 t=tt.,,,r -,õ=õ..õ., 3=:,1 ,4====
0.460 1...., ..... .-, ,,, 1.06 i 16 6.3 [0.51 t,=.r .4 .."6" s''''e =
8 kl ii 0.81 [0.21 i 7 ) i 15 [1]
..". eP
9 It , O. s=0......:=<.i:

t ' L 0"
[001841 In Table 1, 'IC50 values represent the half maximal (50%) inhibitory concentration as determined in the UV-Vis cuvette-based assay in triplicate. bCompound possessed low efficacy, with less than 50% maximal inhibition at 25ILLM inhibitor.
[001851 FIG. 1.B represents the general methodology utilized for the synthesis of compounds listed in Table 2. Towards this end, 5-Amino-2-(naphthalen-l-yl)oxazole-4-carbonitrile 19 was converted to an advanced intermediate (19a), via Sandmeyer reaction by treating with /-butyl nitrite in the presence of copper (II) chloride. Heating the various amines to reflux with intermediate 19a in THF afforded the required analogs 20-26 and 28-31.
Compound 27 was obtained via acetylation of intermediate 19 with acetic anhydride, cat. DMAP.
Table 2. Variations to compound 1. (analogs 19-31).1 ti e'R
Ii \=1 \CN
.........
tC!sij (AM) Compound 0: SD (p.M)1 1 0.20 [0.04]
19 NI=k 25 [
20 MM >30' 21 IN ' 0.12 [0.3]
.22 0.10 [0.31 23 0..1.2 [0.05]
24 1IN ¨"µ""`"'" 0.3 [0.04]
3.0111 26 HIN >40 27 N1-1A 10 [31 29 ti:N=Pti >30"
30 IN.<0 >25 31 .
UN <Nfl >40 1.001861 In Table 2, 0IC50 values represent the half maximal (50%) inhibitory concentration as determined in the UV-Vis cuvefte-based assay in triplicate. bCompound possessed low efficacy, with less than 50% maximal inhibition at 25 uM inhibitor.
Si [001871 The synthesis of compounds 32-40 (Table 3) is outlined in FIG. 1C.
Analogs 32-34 were prepared via chlorination of the intermediates, 32a-34a, with hexachloroethane in the presence of LDA, followed by condensation with methylamine. Amination of 5-chloro-2-(naphthalen-l-yl)thiazole-4-carbonitrile (32b) required microwave heating in a polar solvent, such as 2-propanol, for complete conversion of the starting material. The details of the synthesis of the intermediate, 32a,53'64 is described somewhere else in the Example.
Intermediates 33a and 33b were synthesized following the literature protocols described for similar compounds. The reaction of 4-methy1-3-thiosemicarbazide with 1-naphthoic acid in the presence of EDC
furnished the oxadiazole analog 35, but surprisingly when T3P coupling condition was used, only the thiadiazol.e analog 36 was formed. Synthesis of compound 37 was accomplished utilizing a Cornforth rearrangement of the intermediate 37b, which is prepared from the intermediate 37a by coupling with methylamine as shown in Schem.e 3. The intermediate 37a was prepared according to a known protocol reported to similar compounds in the literature.65 The synthesis of analog 38 from 1-naphthyl boronic acid and the intermediate 38a using classical. Suzuki conditions was met with limited success. 66 The Suzuki coupling reaction was successful using a silica bound DPP-Pd catalyst (SiliaCae-DPP-Pd) under reflux conditions over 24 hours.
Synthesis of the intermediates, 39a-40a, were accomplished using a Pd(PPh3)4-catalyzed microwave assisted regioselective Suzuki coupling between 1-naphthyl boronic acid and 2,6-dichloro pyridine or pyrimidine derivative, respectively. Subsequent condensation of these intermediates with methylamine furnished the desired analogs, 39-40, following HPLC
purification.
Table 3. Variations to compound 1. (analogs 32-40) \
ICse (p.M) Compound 1.1: SD (pM)I
SSS
MIN%
?t-4 0.2010,04:I

NH Me 32 0..55 [0.061 C.34 cl,õN1K4 >40 wAli 34 >40 NI' 14 5,4 [1.81 s 36 $.e >40 . -woo >40 s ...t11MEt 38 1, AO >40 s?iiitsk M-4 >40 1001881 In Table 3, air,50 values represent the half maximal (50%) inhibitory concentration as determined in the UV-Vis cuvette-based assay in triplicate. tompound possessed low efficacy, with less than 50% maximal inhibition at 25 nM inhibitor.
1001891 Results and Discussion 1001901 To investigate structural requirements for optimal 12/15-LOX
inhibition, a SAR
study of the lead molecule was conducted, as shown in Tables 1-3 (1-40).
Initially, the 1-naphthyl group on the left side of the molecule was replaced with various aryl and heterocyclic groups as shown in Table 1. The bioisosteric replacement of 1-naphthyl with 2,3-dichlorophenyl (7) or 3,4-dichlorophenyl (8) groups showed comparable, albeit slightly lower potencies than 1 (IC50 = 0.46 p.M, 0.81 AM and 0.20 p.M, respectively). Replacement with a 2-naphthyl group showed reduced potency (2, IC50> 30 p.M) compared to the 1-naphthyl substitution, which based on space-filling analysis, agreed with the findings observed for dichloro analogs 7 and 8. Several other modifications, including saturated rings or heterocyclic rings at this region (3-6 and 9-18), also resulted in reduced potency. Thus, in general, the 1-naphthyl group appeared to be optimal for 12/15-LOX. inhibition, as variations in size and electrostatics in this region were not well tolerated. Accordingly, the 1-naphthyi group was held constant while other regions of the molecule were explored for further SAR (Tables 2 & 3).
[001911 Having explored modifications to the 1-naphthyl moiety, the next focus was to explore modifications of the N-methyl side chain at the 5-position, as shown in Table 2 (19-31).
Removal of the methyl group drastically reduced potency (19, IC50 = 25 uM) and di-methylation reduced potency as well (20, IC50 > 30 p,M). Modifications of the methyl group with other alkyl substitutions, such as ethyl, n-propyl, n-butyl and n-pentyl groups, were tolerated (21-25), with comparable or even improved potency being observed. However, replacing the methyl group with a branched alkyl group (26) or cycloalkyl groups (30-31) significantly reduced the potency.
Larger groups, such as benzyl (28, IC50> 30 p.M) or phenyl substituted analogs (29, IC50> 30 p.M), also showed diminished 12/15-LOX activity. Overall, these data suggested that the mono-alkylation with straight chain alkyl groups is important for optimal 12/15-LOX
inhibition.
[001921 Finally, attention was turned to optimization of the 1,3-oxazole core and its various substituents (Table 3). Replacing the 1,3-oxazole core with a 1,3-thiazole ring (32, IC50 = 0.55 p.M) resulted in a two-fold decrease in potency. This result could be partly attributed to the difference in size and hardness/softness of the sulfur and oxygen atoms in the thiazole and oxazole rings respectively. The diminished potency trend continued further for the pyridine (39, IC50> 40 AM) and pyrimidine analogs (40, IC50> 40 AM). Attempts to mimic the interaction of the oxazole core were largely unsuccessful with the N-methyl/oxadiazole derivative (35, IC50 =
5.4 AM) or the thiadiazole analog (36, IC50> 40 uM). Interchanging the positions of the 1-naphthyl group and the -NHMe group (analogs 33-34) also led to complete loss of activity. In summary, this chemotype showed very tight SAR and the efforts to make several changes around the lead molecule resulted in similar or lower potency, with exception to analogs 21-23, which showed marginally improved potency.
1001931 Biological Evaluation of ML351 [001941 Concurrently with the SAR investigations against human 12/15-LOX in vitro, the potency of 1 against mouse 12/15-LOX was investigated, utilizing a neuronal cell based assay.
Glutamate-induced oxidative stress67 leads to time-dependent cell death mediated by 12/15-LOX in both immature primary neurons and mouse hippocatnpal. H122 cells.38' 46' 7 Primary neurons require a lengthy isolation procedure and also show relatively modest increases in cell death in this assay, indicating they arc not suited for a high-throughput testing approach. in contrast, H122 cells grow quickly and consistently feature low cell death under control conditions, but a majority of cells die after glutamate treatment (50-100%).
This assay can be adapted to 96-well plates, allowing for moderate to high throughput against the screened.
compounds. The characteristic Z' score used to evaluate the technical quality and suitability as screening tool averaged 0.77 over ten experiments (SD = 0.13; range, 0.49-0.94), where values above 0.5 indicate an excellent assay.71 [001951 To verify that inhibition of oxidative stress-related cell death in HT22 cells is indeed a good measure of the efficacy of a given inhibitor against the mouse 12/15-LOX, the characteristics of cell injury in this model was investigated. Levels of the mouse 12/15-LOX
product, 12-1-1ETE, are known to be increased in the mouse brain following ischemia,34 and they are also elevated by glutamate treatment of 11122 cells.34' 38 HPLC/MS was used to measure 12-HETE secreted to the medium from cells incubated under control conditions or treated with glutamate, as well as co-treated with several compounds with or without inhibitory activity for mouse 12/15-LOX. Following exclusion of one outlier with extremely high 12-HETE (but also high levels of cell death), there was a good correlation between cell death and levels of 12-HETE
secreted; when 12-HETE levels were high, there was increased cell death, when 12-HETE levels were low, there was little cell death (FIG. 2A). Interestingly, levels of 15-HETE were also increased in the medium of glutamate-treated HT22 cells, to a similar extent as 12-HEIE (FIG.
2B). This was surprising because the mouse 12/15-LOX is generally thought to make only minor amounts of 15-HETE, typically less than 20% of the 12-HETE generated.
Nonetheless, 15-HETE production was also blocked by 12/15-LOX inhibitors that protected H122 cells and reduced 12-HETE levels, indicating that both arachidonic acid metabolites were made by the same enzyme.
[001961 Upon testing of compound 1 in 11T22 cells, a dose-dependent protection against oxidative glutamate toxicity (FIG. 3A) was found. To verify the on-target efficacy of 1, 12-HETE secreted from the cells was also measured. The increase in 12-HETE
following glutamate treatment was completely reversed by incubation in the presence of 10 AM of 1 (FIG. 3B).
These results suggest that 1 is capable of reaching its target in the cell and effectively inhibiting the mouse 12/15-LOX, albeit with lower affinity than seen against human 12/15-LOX in vitro (IC50= 200 nM).
[001971 In addition to analogs that were active in vitro against human 12/15-LOX, several inactive analogs (11, 31, 34) were also tested in HT22 cells, with the goal of identifying a good negative control (FIG. 4). Surprisingly, 11 and 31 featured similar protective qualities at 5 AM
concentration, suggesting they are able to inhibit the mouse homologue of 12/15-LOX, even though they are inactive against human 12/15-LOX. In contrast, 5 AM of 34 did not protect HT22 cells, suggesting it is inactive against both human and mouse 12/15-LOX
and is a suitable negative control for 1. These data re-emphasize the importance of screening inhibitors against both. human and mouse 12/15-LOX, since activity against one species does not guarantee activity or inactivity against the other.
[001981 Upon the determination that 1 was potent against both in vitro human 12/15-LOX and ex vivo mouse 12/15-LOX (HT-22 cell assay), the selectivity of a few of the top analogs against related human LOX isozymes (5-LOX, 12-LOX and 15-LOX-2) was then investigated.
Of the four 12/15-LOX inhibitors tested, 1, 7, 8, and 32, all displayed excellent selectivity against all 3 isozymes (IC50 > 50 AM, Table 4). Few compounds reported in the literature have achieved nM
potency towards 12/15-LOX while maintaining excellent selectivity towards other isozymes.
Moreover, none of these analogs displayed inhibition against cycloox.ygenase-1 (COX-1) and/or COX-2 (<10% at 15 AM).
[001991 Mechanistic Investigations of Compound 1 [002001 LOX inhibitors are known to exhibit a variety of inhibitory mechanisms, such as chelative, reductive or competitive. The UV-Vis pseudoperoxidase activity assay was therefore performed on four selected analogs (1, 7, 8, 32, Table 4) to determine if the mechanism was reductive in nature. It was observed that the hydroperoxide product was not degraded, 12/15-LOX was not irreversibly inhibited and there was no elongation of the enzymatic lag phase (data not shown). These data are consistent with a non-reductive inhibitory mechanism. To investigate the nature of inhibition further, steady-state kinetics were performed using compound 1 by monitoring the formation of 15-HpETE as a function of substrate and inhibitor concentration in the presence of 0.01% Triton X-100. Replots of and 1Nn,õ, versus inhibitor concentration yielded linear plots (FIG. 7), with K1 equaling 0.1 +1- 0.002 1.1.M and Ki. equaling 1.2 +/- 0.02 uM. These parameters are defined as the equilibrium constants of dissociation from the catalytic (KO and secondary sites (K1'), respectively. The K1 is in good agreement with the IC50 value and due to the greater than 10-fold difference between Ki and Ki', the secondary site was assume to be the all.osteric site,72'73 which is consistent with previous studies of 12/15-LOX
inhibition.56 Table 4. Selectivity profiling of compound 1 and other top compounds.
12/15- Redox Analog 15-1.0X-26 12-1,0,0 5-11.0X."
Activityb 1 0.02 >100 >100 >50 No 7 0.46 >100 >100 >100 No 8 0.81 >100 >100 >50 No 32 0.55 >100 >40 >50 No 1002011 In Table 4, aIC50values are reported in uM. UV-vis pseudoperoxidase activity assay was performed on all the four selected analogs and no degradation of the hydroperoxide product was observed at 234 nm., indicating a non-reductive inhibitory mechanism.
1002021 In vitro ADME and in vivo PK profile 1002031 The previously reported small molecule inhibitor for 12/15-LOX
inhibition, M1094, demonstrated excellent potency and selectivity but lacked sufficient solubility, cell permeability and microsomal stability (11/2 <2 mins).56 Moreover, this compound possessed an essential ester moiety that could be susceptible to intracellular and plasma esterases, rendering it inactive and possibly limiting its utility in advanced biological models. Thus, the in vitro ADME (Table 5) and in vivo PK properties (Table 6) of 1 represent a vast improvement over the majority of compounds reported previously (vide infra). The majority of the compounds possess the low molecular weight (e.g. 249 Da), and a favorable log D (pH 7.4) between 2-3, which was obtained by Analiza Inc. using their "scaled-down shake flask lipophilicity method", yet most analogs have poor solubility. The aqueous kinetic solubility in PBS buffer (pH 7.4) was determined to be 1.2 gM, which is about 7 times the in vitro IC50. Empirically, a vast improvement in the solubility in the 15-LOX assay buffer was observed (data not shown), which was encouraging and suggests that solubility was not a detrimental factor in the biochemical studies. Importantly, the compound demonstrated favorable PAMPA permeability (passive) and acceptable Caco-2 permeability of >1 (1.5 cm/s-6) with no evidence of efflux (efflux ratio: 0.7) indicating the compound is not susceptible to the action of P-glycoprotein 1 (Pgp), a well-characterized ABC-transporter. Moreover, 1 was stable in various aqueous solutions (pH 2, pH
7.4, pH 9) (described somewhere else in the Example) and mouse plasma (Table 5). In addition, 1 exhibited minimal CYP inhibition of the 2D6 and 3A4 isoforms at 10.3% and 3.5%
inhibition respectively. Microsomal stability appears to be species dependent with 1 possessing moderate stability to rat liver microsomes (18 minutes) while being less stable to mouse liver microsomes (5.5 mins). The compounds were completely stable in the absence of NADPH, suggesting a CYP-mediated degradation. Given the plan to test compound 1 in proof of concept mouse models of stroke, the inventors also obtained in vivo PK data on 1 (Table 6) and found a suitable formulation for 1(10% Solutol, 10% Cremophor EL, 20% PEG4)0 in saline). As anticipated from the microsomal stability studies, I has a relatively fast half-life in both plasma and brain (1112 = I h), with a Cmn of 13.8 gM in plasma and 28.8 gM in brain.
Encouragingly, 1 has a brain/plasma ratio of 2.8, which demonstrates favorable BBB permeability and suggested that this compound was suitable for in vivo proof of concept (POC) models of ischemic stroke (vide infra).
Table 5. ADME profile for Compound 1 Kiftfie CAT 206 CAT 344 /01A.stla is:berm goal Stability Permeability Compound SolubilitY 4 Inhibition bildbitioa 4 'II 2 (131iiiI) õ ....... (10 crotS) (PA) o "JIM fg 01,4411 k 503 i51; 0 :2;
si 2 130E3 r,, 18 5.5 723 1.5 I .2 10.30% 3.50%
(rat) j (mouse) (PAMPA) (Caco-2) 1002041 In Table 5, all experiments were conducted at Pharmaron Inc. "In PBS
buffer (pH
7.4). bRepresents the stability in the presence of NADPH. The probe compound showed no degradation without NADPH present over a 1 hr period. eDextromethorphan was used as the substrate. d Midazolam was used as the substrate.
Table 6. In vivo PK (mouse) at 30 mpk IF' for Compound 1 AITC inf Compoun0_ ,, . .. . . Brain/Plasmab Plasma 1.1 h 0.25 h 13.8 13 2.8 Brain I h 0.5 h 28.8 35.5 1002051 In Table 6, all experiments were conducted at Pharmaron Inc. using male CD1 mice (6-8 weeks of age). Data was collected in triplicate at 8 time points over a 24 h period.
'Formulated as a solution (10% Solutol, 10% Cremophor EL, 20% PEG 400 in saline). bBrain to plasma ratio [ALICk,st(brain)/ AUCksi(plasma)].
(002061 Cell Activity and In vivo efficacy 1002071 After having established the SAR profile against recombinant human 12/15-LOX, determining the efficacy of! in the cellular mouse 12/15-LOX assay, and measuring its in vitro ADME/in vivo PK properties, its efficacy in mouse models of ischemic stroke was then determined. For these initial studies, the permanent focal ischemia model in mice was chosen, which has been shown to mimic the pathophysiological mechanisms following ischemic injury.74 The advantage of this model is that it does not inflict surgical trauma, and has a low mortality compared to other methods. The thrombosis event in the middle cerebral artery (MCA) is induced by topical application of FeCl3 to the intact dura mater, leading to a cortical infarct.
Laser Doppler flowmetry is used to monitor blood flow reduction and infarct size is measured following sacrifice at 24 hours by staining of 1 mm thick brain slices with 2,3,5-triphenylterazolium hydrochloride (TTC). IP administration of! administered 2 hours after the induced ischemia resulted in a ¨30% reduction in infarct size (p < 0.01), demonstrating efficient ncuroprotection in this mouse model of permanent focal ischemia (FIG. 5).
1002081 Conclusion [002091 12/15-LOX contributes to neuronal cell death in oxidative stress mode1s38-44 and is also detrimental to not just neurons, but also to the brain vasculature after stroke,45 via intracellular attack on mitochondria and translocation of the apoptosis-inducing factor (AIF) to the nucleus.46.48 Moreover, the protein level of 12/15-LOX gradually increases over time after experimental stroke,47 providing an opportunity for therapeutic intervention.
Initial efforts to identify a stroke relevant 12/15-LOX inhibitor proved unsuccessful due to its inactivity in the mouse ex vivo model, either due to cellular inactivation or inactivity against mouse 12/15-LOX.56 Fortunately, re-interrogation of the HIS data along with simultaneous cellular mouse 12/15-LOX screening revealed another I 2/15-LOX inhibitor (1), with better "drug-like"
properties and more importantly, mouse ex vivo activity. The initial medicinal chemistry efforts, described herein, provided important SAR. to guide future investigations. The most significant loss of inhibition was observed at the 1-naphthyl position, where almost all substitutions resulted in significant loss in activity, with the exception of its bio-isosteric dichloro analogs (7 and 8), which only showed a slight decrease in activity. Modifications of the methyl group on the N-methyl side chain were not well tolerated either, indicating the straight chain, mono alkylated amine was critical for potency. Moreover, the only substitution tolerated for 1,3 oxazole core ring was the replacement with a 1,3 diazole (32), which resulted in a two-fold decrease in the potency. The cyanide group on the oxazole core was found to be essential, with modifications to this group completely abolishing activity. Subsequent selectivity studies of 1 and related analogs revealed that these compounds have minimal activity towards 5-LOX, 12-LOX, 15-LOX-2, COX-1 and COX-2.
[0021.0j Most importantly, 1 was active in both protecting mouse neuronal cells (HT22) and in reducing the infarct size of stroke-induced mice. This is a significant discovery because identify, ing therapeutically useful specific inhibitors of 12/15-LOX is not trivial due to the differences between the human and mouse 12/15-LOX isozymes. Interestingly, analogs of compound 1(11 and 31) did not inhibit human 12/15-LOX, but protected HT22 cells against oxidative stress. These might work in mice to protect against experimental stroke, yet in human stroke, they would most likely fail, because of their inability to target human 12/15-LOX. The lessons to be drawn from this are that simply because a given inhibitor is selective for a LOX
isoform in one species, its efficacy cannot be readily extrapolated to other species. Moreover, the LOX isotype involved in a given process cannot be reliably inferred from inhibitor experiments without taking into account the species from which the cells were derived.
1002111 In conclusion, compound 1 is the first highly selective 12/15-LOX
inhibitor to be active against human 12/15-LOX and mouse 12/15-LOX, as seen by its effectiveness in the mouse stroke model. This critical quality of 1 is significant because it maintains the human 12/15-LOX selectivity, which is beneficial to a human therapeutic, but is also functional in a mouse stroke model, which is critical to developing its biological efficacy.
Therefore, by using pipeline approach described herein, the inventors have identified a potent and selective inhibitor of human 12/15-LOX that is neuroprotective in a mouse model of stroke and therefore can be utilized in pre-clinical studies to develop it as a potential first-line stroke therapy.
[00212] Experimental Section [00213] General Methods for Chemistry. All air or moisture sensitive reactions were performed under positive pressure of nitrogen with oven-dried glassware.
Anhydrous solvents such as dichloromethaneõKN-dim.ethylforamide (DMF), acetonitrile, diox.an, dimethoxyethane, methanol and triethylamine were purchased from Sigma-Aldrich. Tetrakis and a 50 % of solution of propylphosphonic anhydride (T3M) in DMF or ethyl acetate were purchased from Stem chemicals and used used as such. SiliaCat Heterogeneous Catalyst DPP-Pd (Catalog # R390-100) was purchased from SiliCycle Inc. Preparative purification was performed on a Waters semi-preparative HPLC system using a Phenomenex Luna C18 column (5 micron, 30 x 75 mm) at a flow rate of 45 mUmin. The mobile phase consisted of acetonitrile and water (each containing 0.1% trifluoroacetic acid). A gradient of 10% to 50% acetonitrile over 8 minutes was used during the purification. Fraction collection was triggered by UV
detection (220 nm).
Analytical analysis was performed on an Agilent LC/MS (Agil.ent Technologies, Santa Clara, CA). Method 1: A 7 minute gradient of 4% to 100% Acetonitrile (containing 0.025%
trifluoroacetic acid) in water (containing 0.05% trifluoroacetic acid) was used with an 8 minute run time at a flow rate of 1 mLimin. A Phenomenex Luna C18 column (3 micron, 3 x 75 mm) was used at a temperature of 50 C. Method 2: A 3 minute gradient of 4% to 100% Acetonitrile (containing 0.025% trifluoroacetic acid) in water (containing 0.05%
trifluoroacetic acid) was used with a 4.5 minute run time at a flow rate of 1 mL/min. A Phenomenex Gemini Phenyl column (3 micron, 3 x 100 mm) was used at a temperature of 50 C. Method 3:
Analysis was performed on an Agilent 1290 Infinity Series HPLC. UHPLC Long Gradient Equivalent 4% to 100% acetonitrile (0.05% trifluoroacetic acid) in water over 3.5 minutes run time of 4 minutes with a flow rate of 0.8 mUmin. A Phenomencx Kinctex 1.7 micron C18 column (2.1 x 100 mm) was used at a temperature of 50 C. Purity determination was performed using an Agilent Diode Array Detector for both Method 1, Method 2 and Method 3. Mass determination was performed using an Agilent 6130 mass spectrometer with electrospray ionization in the positive mode. 1H

NMR spectra were recorded on Varian 400 MHz spectrometers. Chemical shifts are reported in ppm with undeuterated solvent (DMSO-d6 at 2.49 ppm) as internal standard for DMSO-d6 solutions. All of the analogs tested in the biological assays have purity greater than 95%, based on both analytical methods. High resolution mass spectrometry was recorded on Agilent 6210 Time-of-Flight LC/MS system. Confirmation of molecular formula was accomplished using electrospray ionization in the positive mode with the Agilent Masshunter software (version B.02).
(00214) Experimental Procedures [002151 General procedure for the preparation of 5-amino-2-substituted oxazole-carbonitrile intermediates (li-1811,19):
[002161 A mixture of carboxylic acid (5.77 mmol, 1 eq.) and 2-aminomalononitrile.Ts0H
(5.77 rnmol, 1 eq.) in ethyl acetate (15 rriL) was added NEt3 (17.32 mmol, 3 eq.) followed by a 50% solution of propylphosphonic anhydride (T3Pc) in ethyl acetate (14.44 mmol, 2.5 eq.). The reaction was allowed to stir at room temperature for 12 h and then diluted with ethyl acetate.
The organic layer was successively washed with water, saturated bicarbonate solution and brine.
The organic layer was then dried with MgSO4, and concentrated under reduced pressure. The crude product was purified on a biotage flash system eluting with 50-100%
ethyl acetate in hexanes containing 0.1 % triethylatnine to provide pure products.
[002171 General procedure for the preparation of 2-substituted-5-(methy1amino)oxazole-4-carbonitrile (1-18).
1002181 A mixture of 5-amino-2-substituted oxazole-4-carbonitrile (1.79 mmol, 1 eq.), paraformaldehyde (0.11 g, 3.57 mmol, 2 eq.) and sodium methoxide (0.096 g, 1.79 mmol, 1 eq.) in methanol (10 mL) was stirred at 65 'V for 1 h until a clear mixture was obtained. The reaction mixrure was cooled to room temperature and sodium borohydride (3.57 mmol, 2 eq.) was added slowly and stirred further at room temperature for 1 h. The crude product was extracted with ethyl acetate and successively washed with water and brine. The ethyl acetate layer was dried with MgSO4, and concentrated under reduced pressure. The crude product was taken up in DMSO and purified on a reversed-phase HPLC system. Characterization data for key compounds is given below.
[002191 5-(Methylamino)-2-(naphthalen-1-ypoxazole-4-carbonitrile (1-ML351):
This compound was prepared starting from 1-naphthoic acid and aminomalononitrile via intermediate 19 following the above general procedure. LC-MS Retention Time: (Method 1) =
6.011 min and (Method 2) = 2.42 mm; IFINMR (400 MHz, DMSO-d6) 6 9.15 (dq, J= 8.7 and 0.9 Hz, 1H), 8.44 (brs, 1H), 8.10 - 7.99 (m, 311), 7.74 - 7.57 (m, 3H), 3.07 - 3.01 (m, 3H); 13C NMR (400 MHz, DMSO-d6) 6 161.9, 161.9, 149.5, 134.1, 134.0, 131.2, 129.2, 128.3, 127.2, 127.2, 127.0, 126.9, 125.8, 125.8, 125.8, 125.7,122.1, 116.5, 116.5, 84.1, 84.1, 84.1, 29.7, 29.7, 29.6; HRMS
(ESI) m/z (M+11)' coded. for C151112N30, 250.0975; found 250.0975.
1002201 5-(Methylamino)-2-(naphthalen-2-y1)oxazole-4-earbonitrile TFA (2). LC-MS
Retention Time: (Method 3) = 2.496 min and (Method 2) = 3.556 min; IFI NMR
(400 MHz, DMSO-d6) 6 8.41 (d, J= 5.0 Hz, 1H), 8.34 (d, J = 1.7 Hz, III), 8.09 -8.00 (m, 211), 7.99 - 7.89 (m, 2H), 7.63 - 7.56 (m, 211), 3.07 - 3.02 (m, 3H); FIRMS (ESI) ink (M+II)+
calcd. for CI5H12N30, 250.0975; found 250.0980.
1002211 2-(lsoquinolin-5-y1)-5-(metbyla rnino)ourcole-4-carbonitrile TFA (3).
LC-MS
Retention Time: (Method 3) = 1.228 min and (Method 2) = 2.466 min; III NMR
(400 MHz, DMSO-4) 6 9.49 (s, 1H), 9.04 (d, J= 6.2 Hz, 1H), 8.68 (d, J= 6.2 Hz, 1H), 8.55 (q, J= 4.8 Hz, 1F1), 8.31 (d, J=. 7.7 Hz, 2H), 7.85 (t, J= 7.8 Hz, 1I1), 3.05 (d, = 4.9 Hz, 31f); FIRMS (ESI) mlz (M-I-H)-1- calcd. for Ci4HIIN40, 251.0927; found 251.0918.
1002221 5-(Methylamino)-2-(quinolin-5-y1)oxazole-4-carbonitrile TEA (4). LC-MS

Retention Time: (Method 1) = 3.621 min and (Method 2) = 2.582 min; NMR (400 MHz, DMSO-d6) 6 9.61 9.53 (m, 1H), 9.01 (dd, J = 4.2, 1.7 Hz, 1H), 8.53 (q,J - 4.9 Hz, 1H), 8.20 --8.08 (m, 2H), 7.94 - 7.83 (m, 111), 7.73 (dd, .1= 8.7, 4.2 Hz, 111), 3.05 (d, .1= 4.9 Hz, 3H);
HRMS (ESI) miz (M+H)+ calcd. for CHIIIIN40, 251.0927; found 251.0938.
1002231 2-(Isoquinolin-6-y1)-5-(methylandno)oxazole-4-carbonitrile TFA (5). LC-MS
Retention Time: (Method 1) = 3.259 min and (Method 2) = 2.534 min; 1H NMR (400 MHz, DMSO-4) ö 9.50 (s, 1H), 8.61 (dd, .1=5.5, 3.8 Hz, 2H), 8.45 (d, J - 1.6 Hz, 1H), 8.33 (d, J=
8.6 Hz, 1H), 8.20 - 8.11 (m, 2H), 3.06 (d,./ = 4.9 Hz, 3H); HRMS (ESI) tn/z (M+H)+ calcd. for CI4HiiN40, 251.0927; found 251.0928.
1002241 5-(Methylamino)-2-(quinoxalin-5-y1)oxazo1e-4-carbonitrile TFA (6). LC-MS
Retention Time: (Method 3) = 1.862 mm and (Method 2) = 2.95 min; NMR (400 MHz, DMSO-d6) 6 9.04 - 8.96 (m, 2H), 8.58 (d, J= 5.0 Hz, 111), 8.37 (d, J= 1.8 Hz, 111), 8.29 - 8.17 (m, 211), 3.05 (dd, J= 5.0, 0.6 Hz, 113); HRMS (ESI) m/z (M+H)+ calcd. for C, 31110N50, 252.088; found 252.0883.

[002251 2-(2,3-Dichloropheny1)-5-(methylamino)oxazole-4-carbonitrile TFA (7).
This compound was prepared starting from 2,3-benzoic acid and aminomalononitrile via intermediate 7i following the above general procedure. LC-MS Retention Time: (Method 3) =
2.639 min and (Method 2) = 3.559 min; IFINMR (400 MHz, Di SO-d6) 6 8.50 (q, J= 4.9 Hz, 1H), 7.83 (ddd, J
=7.9, 1.5, 0.4 Hz, 1H), 7.78 (ddd, J= 8.1, 1.6, 0.4 Hz, 1H), 7.54 ¨ 7.46 (m, 1H), 2.99 (d, ./ = 4.9 Hz, 3H); HRMS (ESI) raiz (M+11)1 calcd. for C1111802N30, 269.0039; found 267.0031.
[002261 2-(3,4-Dich1oropheny1)-5-(methylamino)oxazole-4-carbonitrile TFA (8).
This compound was prepared starting from 3,4-benzoic acid and aminomalononitrile via intermediate 81 following the above general procedure. LC-MS Retention Time: (Method 2) =
3.737 min;
NMR (400 MHz, DMS046) 6 8.48 (q, J= 4.9 Hz, 1H), 7.95 (td, J= 1.8, 0.7 Hz, 1I1), 7.80 ¨
7.72 (m, 2H), 3.09 ¨ 2.91 (m, 311); HRMS (ESI) m/z (M+H).- calcd. for CI
1118Cl2N30, 269.0039;
found 267.0043.
[002271 2-(2,3-Dihydrobenzo[b][1,41dioxin-6-y1)-5-(methylamino)oxazole-4-carbonitrile TFA (9). LC-MS Retention Time: (Method 1) = 4.905 min and (Method 2) = 3.323 rain; 111 NMR (400 MHz, DMSO-d6) 6 8.24 (q,..1= 4.9 Hz, 111), 7.34 7.15 (m, 211), 6.97 (dd, J::: 8.5, 0.4 Hz, 1H), 4.29 (d, f= 1.0 Hz, 4H), 2.96 (d, J= 4.9 Hz, 3H); HRMS (ESI) m/z (M-FH). calcd.
for C13E1121\1303, 258.873; found 258.875.
[002281 2-(Benzo[d][1,31dioxol-5-y1)-5-(methylamino)oxazole-4-carbonitrile TFA
(10).
LC-MS Retention Time: (Method 3) = 2.019 min and (Method 2) = 3.338 min; 1H
NMR (400 MHz, DMSO-d6) 6 8.24 (q, J = 5.0 Hz, 1H), 7.45 ¨ 7.18 (m, 2H), 7.03 (dt, J =
8.1, 0.5 Hz, 1H), 6.10 (d, J= 0.6 Hz, 211), 2.97 (d, .1= 5.0 Hz, 3H); HRMS (ESI) m/z (M+H)+
calcd. for Ci211i0N303, 244.0717; found, 244.0712.
1002291 2-(11,1'-Bipheny11-4-y1)-5-(methylamino)oxazole-4-carbonitrile TFA
(11). This compound was prepared starting from 4-phenylbenzoic acid and aminomalononitrile via intermediate 111 following the above general procedure. LC-MS Retention Time:
(Method 1) =
6.215 min and (Method 2) = 3.708 min; IFINMR (400 MHz, DMSO-d6) 6 8.38 (q, J=
4.9 Hz, 1H), 7.92 ¨ 7.79 (m, 4H), 7.76 ¨ 7.68 (m, 2H), 7.53 ¨ 7.46 (m, 211), 7.43 ¨
7.37 (m, 111), 3.01 (dd, J= 4.8, 0.6 Hz, 31); HRMS (ESI) m/z (M+H)+ calcd. for C171114N30, 276.1131; found 276.1143.
1002301 2-(1H-indo1-6-y1)-5-(methylamino)oxazole-4-carbonitrile TFA (13). LC-MS
Retention Time: (Method 3) = 2.004 min and (Method 2) = 3.308 min; 1H NMR (400 MHz, DMSO-d6) 6 11.38 (s, 111), 8.22 (q, J= 4.9 Hz, 1H), 7.83 (dq, J= 1.4, 0.7 Hz, 1H), 7.63 (dt, J =
8.3, 0.7 Hz, 1H), 7.56 -7.41 (m, 2H), 6.49 (ddd, J= 3.0, 1.9, 0.9 Hz, 1H), 3.00 (d, J= 5.0 Hz, 3H); HRMS (ES1) m/z (M+H) calcd. for ColliiN40, 239.0927; found 239.0934.
1002311 2-(1H-indo1-3-y1)-5-(methylamino)oxazole-4-varbonitrile TFA (14). LC-MS
Retention Time: (Method 3) = 1.938 min and (Method 2) = 3.236 min; 11-1 NMR
(400 MHz, DMSO-d6) 8 11.73 (d, J= 13.4 Hz, 111), 8.10 - 7.95 (m, 211), 7.89 (dd, J=
10.7, 2.8 Hz. 111), 7.52- 7.44 (m, III), 7.26 - 7.12 (m, 211), 2.99 (d, J= 5.0 Hz, 3H); 'FIRMS
(ES1) m/z (M+II)+
calcd. for Ci3Hi1N40, 239.0927; found 239.0923.
[002321 Synthesis of 5-(Azetidin-3-ylamino)-2-(naphthalen-1-ypoxazole-4-carbonitrile TFA (31). A mixture of 5-chloro-2-(naphthalen-1-yDoxazole-4-carbortitrile (19a) (0.25 mmol, 1 eq.) and 1-Boc-3-(amino)azetidine (1 mmol, 4 eq) in THF was heated to reflux for 0.5 h. The reaction mixture was then cooled and the solvent was removed by forced air.
The crude product was taken up in DMSO and purified via reversed phase preparative HPLC. This pure product was suspended in dichloromethane (2 mL) and treated with TFA (0.5 mL). After stirring for 0.5 h at room temperature, the volatiles were removed by forced air. The crude product was taken up in DMSO and purified on a preparative HPLC to give pure product (31) as TFA
salt. LC-MS
Retention Time: (Method I) = 4.078 min and t2 (Method 2) =3.812 min; 1H NMR
(400 MHz, DMSO-d6) 6 9.29 (s, I H), 9.12 (d, J= 8.6 Hz, 1H), 8.90 (s, 1H), 8.14- 8.01 (m, 2H), 7.75 - 7.59 (m, 3H), 4.77 (p, - 7.0, 6.0 Hz, I H), 4.32 4.18 (m, 2H), 4.18 4.08 (m, 2H);
FIRMS (ESD
m/z (M+H) calcd. for CI7H15N40, 291.1252; found 291.125.
1002331 Synthesis of (methylaraino)-2-(naphthalen-1-yOthiazole-4-carbonitrile TFA (32).
A mixture of 2-bromothiazole-4-carbonitrile (1.85 g, 9.79 mmol, 1 eq.), naphthalen-l-ylboronic acid (2.52 g, 14.68 mmol, 1.5 eq.), 2 M sodium carbonate (12.23 niL, 24.47 mmol, 2.5 eq.) and Pd(PPh3)4 (1.11 g, 0.979 mmol, 10 mol%) in dimethoxy ethane (20 mL) was degassed with argon then heated under microwave for 45 minutes at 150 C. The reaction mixture was concentrated and taken up in dichloromethane then stirred with palladium scavenger resin and filtered through celite. The crude product obtained after evaporating the solvent was purified on a biotaget flash system eluting with 10% ethyl acetate in hexanes to obtain 1.52 g (Yield: 66 %) of 2-(naphthalen-1-yOthiazole-4-carbonitrile (32a) as white solid.
[002341 A solution of the above 2-(naphthalen-1-yl)thiazole-4-carbonitrile 32a (0.5 g, 2.12 mmol, 1 eq.) in THF (10 mL) was added a 2 M THF solution of LDA (1.16 mL, 2.33 mmol, 1.1 eq.) dropwise at -78 C. After stirring for 30 minutes at -78 C, perchloroethane (0.551 g, 2.33 mmol, 1.1 eq) was added in one portion and allowed to warm to room temperature over 4 h. The reaction was then quenched with saturated ammonium chloride and extracted with ethyl acetate.
The organic layer was washed with water and brine. The ethyl acetate layer was subsequently dried over MgSO4 and filtered. The crude product obtained after concentrating under diminished pressure was purified on a Biotage flash system eluting with 50% C112C12 in hexanes to furnish 4.5 g (Yield: 79%) of 5-chloro-2-(naphthalen-l-yOthiazole-4-carbonitrile 32b as a white solid.
111 NMR (400 MHz, DMSO-d6) 6 8.67 (d, J= 8.4 Hz, I H), 8.20 (d, J= 8.3 Hz, III), 8.13 - 8.05 (m, 111), 7.94 (dd, J = 7.3, 1.3 Hz, 1H), 7.69 (dddd, J= 14.7, 9.3, 6.9, 1.7 Hz, 311).
[002351 A mixture of 5-chloro-2-(naphthalen-l-yl)thiazole-4-carbonitrile 32b (0.2 g, 0.739 mmol, I eq.) and a 2 M THE solution of methylamine (1.9 mi., 3.69 mmol, 5 eq) in 2-propanol (1 mL) was heated via microwave for 30 min at 120 'C. The crude product obtained after evaporation of the solvent was taken up in DMSO and purified by reversed phase preparative HPLC to obtain 5-(methylamino)-2-(naphthalen-l-yOthiazole-4-calbonitrile (32) as TEA salt.
LC-MS Retention Time: (Method 3) = 2.814 min and (Method 2) = 3.756 min; 1H
NMR (400 MHz, DMSO-d6) 6 8.88 (ddt, = 8.5, 1.4, 0.8 Hz, 1H), 8.17 (q, .J= 4.7 Hz, 1H), 8.07 7.98 (m, 21-1), 7.77 (dd, J = 7.2, 1.2 Hz, 1H), 7.71 - 7.53 (m, 3H), 3.01 (d, J= 4.7 Hz, 3H); HRMS (ES1) m/z (M-1-H) calcd. for Ci5Hi2N3S, 266.0754; found 266.0746.
(002361 Synthesis of N-methy1-5-(naphthalen-1-y1)oxazol-2-amine (34). A
solution of 5-(naphthalen-1 -yl)oxazole-4-carbonitrile 34a (0.9 g, 4.09 mmol, 1 eq.) in THE
(15 mL) was added a 1 M THE solution of LHMDS (4.5 mL, 4.5 mmol, 1.1 eq.) dropwise at -78 C.
The reaction mixture was stirred for 30 minutes at -78 C then perchloroethane (1.064 g, 4.5 mmol, 1.1 eq.) was added in one portion and allowed to reach room temperature over 4 h. The reaction mixture was quenched with saturated ammonium chloride and extracted with ethyl acetate. The organic layer was washed with water and brine then dried over magnesium sulfate, filtered and concentrated under diminished pressure. The crude product was purified on a Biotage flash system eluting with 10% ethyl acetate in hexanes to furnish 0.75 g (Yield: 72 %) of 2-chloro-5-(naphthalen-1-ypoxazole-4-carbonitrilc 34b as white solid.
[002371 A solution of 2-chloro-5-(naphthalen-l-ypoxazole-4-carbonitrile 34b (0.1 g, 0.393 mmol, 1 eq.) in THE (1 mL) was added a 2 M solution of methanamine in THE (1.0 mL, 1.98 mmol, 5 eq.) and stirred at 65 C for 1 h in a sealed tube. The solvent was removed by forced air to obtain a crude solid. This crude product was taken up in DMSO and purified on a preparative HPLC to obtain pure product (34) as TFA salt. LC-MS Retention Time: (Method 1) = 5.509 min and (Method 2) = 3.498 min; NMR (400 MHz, DMSO-d6) 6 8.17 -7.96 (m, 4H), 7.81 (dd, = 7.3, 1.3 Hz, 1H), 7.71 - 7.59 (m, 3H), 2.88 (d, J = 4.8 Hz, 31); HRMS (ES!) m/z (M+H)+
calcd. for Ci5fluN30, 250.0975; found 250.0979.
[00238] General procedure for the synthesis of compounds 20-26 and 28-31 (FIG.
1D) 1002391 A suspension of copper(II) chloride (13.10 g, 97 mrnol, 2.05 eq.) in acetonitrile (200 naL) was added t-butyl nitrite (13.82 mL, 105 mmol, 2.2 eq.) followed by 5-amino-2-(naphthalen-1 -yl)oxazole-4-carbonitrile (11.18 g, 47.5 mmol, 1 eq.) in portions at room temperature. The reaction mixture was stirred for 3 h then concentrated and extracted with ethyl acetate. The organic layer was successively washed with 1% HC1, water and brine. The organic layer was dried over MgSO4, filtered and concentrated under diminished pressure. The crude product was purified on a biotagee flash system eluting with 40% CH2C12 in hexanes to furnish 4.2 g (Yield: 35%) of 5-chloro-2-(naphtbalen-1-y1)oxazole-4-carbonitrile (19a) as white solid.
NMR (400 MHz, DMSO-d6) 6 8.99 (dq, J --= 8.6, 0.9 Hz, 1H), 8.27 --- 8.18 (m, 211), 8.09 (ddt, = 8.1, 1.3, 0.6 Hz, 1H), 7.79 - 7.63 (m, 3H).
1002401 A mixture of 5-chloro-2-(naphthalen-l-yl)oxazole-4-carbonitrile (19a) (0.25 mmol, 1 eq.) and appropriate amine (10 eq) in THF was heated to reflux for 0.5 h. The reaction mixture was then cooled and the solvent was removed by forced air. The crude product was taken up in DMSO and purified via reversed phase preparative HPLC to obtain pure products as TFA salts.
1002411 5-(Ethylamino)-2-(naphthalen-1-yl)oxazole-4-carbonitrile TFA (21). LC-MS
Retention Time: (Method 1) = 6.278 min and (Method 2) = 3.724 min; Ill NMR
(400 MHz, DMSO-d6)6 9.14 (ddq, .1= 8.6, 1.3, 0.7 Hz, 1H), 8.55 (t, .1=5.6 Hz, 1H), 8.12 -8.00 (m, 3H), 7.75 - 7.57 (m, 3H), 3.47 - 3.41 (m, 2H), 1.26 (td, 7.2, 0.6 Hz, 3H); HRMS
(ES1) m/z (M+H)# Med. for C161-114N30, 264.1131; found 264.113.
[00242] 2-(Naphthalen-1-y1)-54propy1amino)oxazole-4-carbonitrile TFA (22). LC-MS
Retention Time: (Method 1) = 6.586 mm and (Method 2) = 3.819 min; 1H NMR (400 MHz, DMSO-%)6 9.15 (ddt, J= 9.1, 1.3,0.7 Hz, 1H), 8.59 (t, J= 5.8 Hz, 111), 8.12 -7.98 (m, 3H), 7.75 -7.57 (m, 3H), 3.38 - 3.35 (m, 2H), 1.71 - 1.60 (m, 2H), 0.96 (td, J=
7.4, 0.5 Hz, 3H);
HRMS (ESI) m/z (M+H)- calcd. for Ci7Hi6N30, 278.1288; found 278.1289.

[002431 5-(Buty1amino)-2-(naphtha1en-1-371)oxazo1e-4-carbonitrile TFA (23). LC-MS
Retention Time: (Method 3) = 2.496 mm and (Method 2) = 3.909 min; 'H NMR (400 MHz, DMSO-d6) 6 9.18 - 9.11 (m, 1H), 8.57 (t, .1= 5.9 Hz, 1H), 8.10 - 7.99 (m, 3H), 7.74- 7.58 (m, 3H), 3.45 -3.38 (m, 2H), 1.68 - 1.56 (m, 2H), 1.48- 1.33 (m, 2H), 0.94 (t, J=
7.4 Hz, 3H);
HRMS (ESI) m/z (M+H) calcd. for C181-118N:10, 292.1444; found 292.1453.
[002441 2-(Naphthalen4-y1)-5-(pentylamino)oxazole-4-carbonitrile TFA (24). LC-MS
Retention Time: (Method 3) = 2.496 mm and (Method 2) = 3.909 min; NMR (400 MHz, DMSO-d6) 6 9.15 (ddt, J= 8.7, 1.4,0.8 Hz, 111), 8.57 (t, J= 5.8 Hz, 111), 8.10 - 7.96 (m, 310, 7.73 - 7.52 (m, 311), 3.43 -3.30 (m, 2H), 1.71 - 1.58 (m, 211), 1.45- 1.26 (m, 4H), 0.96 - 0.82 (m, 3H); FIRMS (ESI) m/z (M+H)i- calcd. for Ci9H20N30, 306.1601; found 306.1609.
[00245] 5-(Allylamino)-2-(naphthallen-1-y1)oxazo1e-4-carbonitrile TFA (25). LC-MS
Retention Time: (Method 2):::: 3.534 min; 1H NMR (400 MHz, DMSO-d6) 6 10.30 (s, 1H), 8.26 -8.12 (m, 1H), 8.13 - 7.95 (m, 111), 7.78 (dd, J= 7.1, 1.2 Hz, 111), 7.74 -7.54 (m, 411), 5.99 (ddtõI= 17.2, 10.2, 7.2 Hz, 1H), 5.63 - 5.47 (m, 2H), 3.21 (dt, J= 7.3, 1.1 Hz, 2H); HRMS
(ES1) m/z (M+H)F calcd. for Ci7H14N30, 276.1131; found 276.1142.
[002461 5-(11sopropylamino)-2-(naphthalen-1.-A)oxazole-4-carbonitrile TFA
(26). LC-MS
Retention Time: (Method 1) = 6.553 min and (Method 2) = 3.82 min; 1H NMR (400 MHz, DMSO-do) 6 9.15 (dq, J= 8.6, 0.9 Hz, 1H), 8.51 (d, J= 7.9 Hz, 1H), 8.11 -7.96 (m, 3H), 7.74 -7.58 (m, 3H), 3.91 (dq, J= 7.8, 6.4 Hz, 1H), 1.29 (d, J- 6.5 Hz, 6H); HRMS
(ESL) m/z (M +HY
calcd. for C,7ffiNI30, 278.1288; found 278.1301.
1002471 5-(Benzylamino)-2-(naphthalen-1-yl)oxazole-4-carbonitrile TFA (28). LC-MS
Retention Time: (Method 3) = 2.975 min and t2 (Method 2) = 3.857 min; 'H NMR
(400 MHz, DMSO-d6) 6 9.14(t, .1=6.2 Hz, 1H), 9.10 -9.04 (m, 1H), 8.10 -7.96 (m, 3H), 7.71 -7.54 (m, 3H), 7.49 -7.35 (m, 4H), 7.34 - 7.25 (m, In), 4.61 (d, J= 6.2 Hz, 2H); HRMS
(ES1) m/z (M+H)4 calcd. for C21Hi6N30, 326.1288; found 326.1295.
[002481 2-(Naphtha1en-1-y1)-5-(pheny1amino)oxazo1e-4-carbonitrile TFA (29). LC-MS
Retention Time: (Method 1) = 6.759 mm and (Method 2) = 3.875 min; 1H NMR (400 MHz, DMSO-d6) 6 9.00- 8.98 (m, 1H), 8.25 - 8.25 (m, 2H), 8.15 - 8.02 (m, 3H), 7.80 -7.60 (m, 51), 7.47 - 7.37 (m, 2H); HRMS (ES!) m/z (M+Na) calcd. for C201114N30, 334.0955;
found 334.0951.

[002491 2-(Naphtha1en-1-yI)-5-(oxetan-3-ylamino)oxazole-4-carbonitrile TFA
(30). LC-MS Retention Time: (Method 3) = 2.484 min and (Method 2) = 3.405 min; IFINMR
(400 MHz, DMSO-d6) 8 9.31 (d, J = 6.1 Hz, 1H), 9.12 (dt, J = 8.9, 1.0 Hz, 1H), 8.14 -7.98 (m, 3H), 7.74 -7.58 (m, 3H), 4.95 - 4.82 (m, 31), 4.69 -4.61 (m, 21); FIRMS (ESI) rn/z (M+H)--calcd. for C17111 4N302, 292.1081; found 292.1088.
[002501 N-methyl-5-(naphthalen-l-y1)oxazol-2-amine (33). For synthesis scheme, see FIG.
IE. A solution of 5-(naphthalen-1-yl)oxazole 33a (0.5 g, 2.56 mmol, 1 eq.) in THF (15 mL) was added a I M TIE solution of LIIMDS (2.82 mL, 2.82 mrnol, 1.1 eq.) dropwise at -78 C. The reaction mixture was stirred for 30 minutes at -78 C then perchloroethane (0.667 g, 2.82 mmol, 1.1 eq.) was added in one portion and allowed to reach room temperature over 4 h. The reaction mixture was quenched with saturated ammonium chloride and extracted with ethyl acetate. The organic layer was washed with water and brine then dried over magnesium sulfate, filtered and concentrated under diminished pressure. The crude product was purified on a Biotage flash system eluting with 10% ethyl acetate in hexanes to furnish 0.54 g (Yield: 92 %) of 2-chloro-5-(naphthalen-l-yl)oxazole 33b as white solid. A solution of 2-chloro-5-(naphthalen- 1-yl)oxazole 33b (0.1 g, 0.435 mmol, 1 eq.) in THF (I mL) was added a 2 M solution of methanamine in THF
(1.09 mL, 2.18 mmol, 5 eq.) and stirred at 65 C for 1 h in a sealed tube. The solvent was removed by forced air to obtain a crude solid. This crude product was taken up in DMSO and purified on a preparative HPLC to obtain N-methy1-5-(naphthalen-1-ypoxazo1-2-amine (33) as TFA salt. LC-MS Retention Time: (Method 3) = 1.593 min and (Method 2) = 2.831 min; Ill NMR (400 MHz, DMS046) 8 8.64 (s, 111), 8.32 - 8.23 (m, 1H), 8.05 -7.93 (m, 2H), 7.74 -7.50 (m, 6H), 2.95 (s, 3H); HRMS (ESI)m/z (M+Hr calcd. for Ci4H13N20, 225.1022; found 225.1032.
[002511 N-methy1-5-(naphthalen-I-y1)-1,3,4-oxadiazol-2-amine 05). For synthesis scheme, see FIG. IF. A mixture of 1-naphthoic acid (0.2 g, 1.16 mmol, 1 eq.), N-methylhydrazinecarbothioamide (0.122 g, 1.16 mmol, 1 eq.) and EDC (0.668 g, 3.48 mmol, 3 eq.) in DMF (3 mL) was stirred at room temperature for 12 h. The reaction mixture was filtered and purified on a HPLC. LC-MS Retention Time: (Method 3) = 1.905 min and (Method 2) =
3.048 min; IFINMR (400 MHz, DMSO-d6) ö 9.11 (ddq, J= 8.7, 1.5, 0.7 Hz, 1H), 8.14 - 7.93 (m, 311), 7.78 -7.50 (m, 411), 2.92 (dd, J= 4.9, 0.6 Hz, 311); HRMS (ESI) miz (M+H).' calcd. for C1 31112N30, 226.0975; found 226.0976.

[002521 N-methyl-5-(naphthalen-1-y1)-1,3,4-thiadiazol-2-amine (36). For synthesis scheme, see FIG. 1G. A solution of 1-naphthoic acid (0.1 g, 0.581 mmol, 1 eq.), N-methylhydrazinecarbothioamide (0.061 g, 0.581 mmol, 1 eq.) in DMF (1 mL) was added triethylamine (0.24 mL, 1.74 mmol, 3 eq.) followed by a 50% solution of propylphosphonic anhydride (T3P) (0.92 g, 1.45 mmol, 2.5 eq.) dropwise. The reaction mixture was stirred at room temperature for 6 h then filtered through a celite plug. The crude product was purified via reversed phase preparative HPLC. LC-MS Retention Time: (Method 3) = 1.852 min and (Method 2) = 2.972 min; 111 NMR (400 MHz, DMSO-d6) 6 8.81 -8.74 (m, 1H), 8.09 -7.94 (m, 3H), 7.74 (dd, J = 7.2, 1.2 Hz, III), 7.68 - 7.56 (m, 3I1), 2.98 (d, J = 4.1 Hz, 3H); FIRMS (ESI) m/z (M+H)+ calcd. for CI3H.12N3S, 242.0746; found 242.0751.
[002531 Methyl 5-(methylamino)-2-(naphthalen-1-3/1)oxazole-4-carboxylate 07).
For synthesis scheme, see FIG. ill. A suspension of 5-methoxy-2-(naphthalen-1-ypoxazole-4-carboxylic acid 37a (3.6 g, 13.37 mmol, 1 eq.) in dichloromethane (50 mi.) was added few drops of DMF and cooled in an ice bath. To the cold suspension was added oxalyl chloride (1.8 mL, 20.06 mmol, 1.5 eq.) dropwise. The reaction mixture was stirred at room temperature for 2 h.
Excess solvent was evaporated under diminished pressure and the product was dried under vacuum. The crude solid was re-dissolved in dichloromethane (25 mL) and a 2 M
solution of methanamine in THF (8 mL, 16.04 mmol) was added followed by triethyl amine (2.80 mL, 20.06 mmol) upon cooling. The reaction mixture was stirred at room temperature for 1 h then concentrated and purified on a Biotage flash system eluting with 50% ethyl acetate in hexanes to furnish 2.2 g (Yield: 58 %) of 5-methoxy-N-methyl-2-(naphthalen-1 -yl)oxazole-4-carboxamide (37b) as yellow solid. A portion of this yellow solid (0.25 g, 0.886 nunoD was heated to reflux in toluene over 12 h. After completion of the reaction, the solvent was removed by forced air. The crude product was taken up in DMSO (2 mL) and purified via reversed phase HPLC. LC-MS Retention Time: (Method 1) = 5.947 mm and (Method 2) = 3.647 min;
IFINMR
(400 MHz, DMSO-d6) 6 9.33 (ddq, J= 8.7, 1.4, 0.7 Hz, 1H), 8.11 (dt, J= 7.5õ
0.8 Hz, 1H), 8.06 -7.98 (m, 2H), 7.73 -7.67 (m, 1H), 7.66 -7.57 (m, 2H), 7.53 (q. J= 4.9 Hz, 1H), 3.78 (d, J=
0.6 Hz, 3H), 3.09 (dd. J = 5.0, 0.6 Hz, 3H); HRMS (ESI) m/z (M+H) calcd. for Ci6Hi5N203, 283.1077; found 283.1085.
[002541 Methyl 5-(methylamino)-2-(naphthalen-1-yl)thiazole-4-earboxylate (38).
For synthesis scheme, see FIG. 1I. To a suspension of methyl 2-bromo-5-(methy1arnino)thiazo1e-4-carboxylate (1.55 g, 6.17 mmol, 1 eq.), naphthalen-l-ylboronic acid (2.12 g,
12.35 mmol, 2 eq.), SiliaCat-DPP-Pd (2.0 g, 0.309 mmol, 5 mol%) (purchased from silicycle, catalog # R390-100) in dimethoxyethane (30 mL) was added a 2 M solution of sodium carbonate in water (9.3 mL, 18.52 mmol, 3 eq.). The reaction mixture was bubbled with argon for 5 minutes and then stirred at 105 C for 24 h. The reaction mixture was concentrated and taken up in dichloromethane and stirred with palladium scavenger resin. After filtering through Mite and removal of the solvent, a portion of the crude product was purified on HPLC. LC-MS Retention Time:
(Method 3) = 2.793 min and (Method 2) = 3.781 min; III NMR (400 MHz, DMSO-d6) 6 9.01 (dd, J= 8.2, 1.6 Hz, 111), 8.08 -7.97 (m, 2H), 7.88 -7.74 (m, 211), 7.72- 7.53 (m., 311), 3.82 (s, 3I1), 3.04 (d, J= 4.9 Hz, 3I1); HRMS (ESI) m/z (M+II)4 calcd. for Ci6H15N202S 299.0849; found 299.0841.
[002551 2-(Methylarnino)-6-(naphthalen-l-Anicotinonitrile 09). For synthesis scheme, see FIG. U. A mixture of 2,6-dichloronicotinonitrile (0.500g. 2.89 mmol, 1 eq.), naphthalen-1 -ylboronic acid (0.522 g, 3.03 mmol, 1.05 eq.) and a solution of sodium carbonate in 3 mi. of water (0.680 g, 8.09 mmol, 2.8 eq.) in dioxane (12 rnL) was bubbled with argon for 5 minutes.
Pd(PP113)4 (0.167 g, 0.145 mmol, 5 mol /0) was then added and heated to reflux under nitrogen for 3 h. The reaction mixture was then stirred with palladium scavenger resin and filtered through celite. The filtrate was concentrated and purified on a Biotaget, flash system eluting with 20% ethyl acetate in hexanes to give 0.45 g (Yield: 59 10) of 39a. The above 2-chloro-6-(naphthalen-1-yDnicotinonitrile (0.12 g, 0.453 mmol) (39a) and a 2 M THF
solution of methanamine (1.13 ml, 2.27 mmol) in 2-iPrOH (1 mL) was heated in a MW for 30 min at 120 (t. The crude product obtained upon concentration was taken up in DMSO and purified via reversed phase HPLC to obtain pure 39 as a TEA salt. LC-MS Retention Time:
(Method 3) =
1.803 min and (Method 2) = 3.796 min; 'H NMR (400 MHz, DMSO-4) 8 8.26 - 8.17 (m, 1H), 8.15 - 7.92 (m, 3H), 7.76 - 7.48 (m, 41-1), 7.25 (q, J = 4.6 Hz, 1H), 6.91 (d, J = 7.8 Hz, 1H), 2.88 (d, .1= 4.5 Hz, 3H); HRMS (ESI) m/z (M+H) calcd. for Ci7H141=13, 260.1182;
found 260.1172.
[002561 N-methyl-4-(naphthalen-1-Apyrimidin-2-amine (40). This compound was synthesized following the protocol used for compound 39 to obtain pure product 40 as a TEA salt after HPLC purification. LC-MS Retention Time: (Method 3) = 1.803 min and (Method 2) =
2.759 min; NMR (400 MHz, DMSO-d6) ö 8.45 (d, J= 5.2 Hz, 1H), 8.25 (m, 1H), 8.04 (ddd, J
= 15.4, 7.7, 2.2 Hz, 2H), 7.71 -7.51 (m, 4H), 6.92 (d, J= 5.2 Hz, 1H), 2.88 (s, 3H); HRMS
(ESI) m/z (M+H)+ calcd. for C151114N3, 236.1189; found 236.1182.

[002571 Materials and Methods (00258) Materials: Different commercial fatty acids as lipoxygenase substrates, were purchased from Nu Chek Prep, Inc. (MN, USA). The fatty acids were further re-purified using a Higgins HAISIL column (5 p.m. 250 X 10 m.m) C-18 column. An isocratic elution of 85%
solvent A (99.9% methanol and 0.1% acetic acid): 15% solvent B (99.9% water and 0.1% acetic acid) was used to purify all the fatty acids. Post purification, the fatty acids were stored at -80 C
for a maximum of 6 months. Lipoxygenase product 13-(S)-HPODE was generated by reacting the linoleic acid (LA) with soybean LOX-1. The product generation protocol involved reacting 50 gM substrate in 500 mI, of 100 mM Borate buffer pH 9.2 with soybean LOX-1.
A. small sample from the big reaction was monitored on the UV-Vis spectrometer till complete turnover.
The products were then extracted using dichloromethane, reduced with trimethylphosphite, evaporated to dryness and reconstituted in methanol. The products were HPLC
purified using an isocratic elution of 75% A (99.9% methanol and 0.1% acetic acid): 25% B (99.9%
water and 0.1% acetic acid). The products were tested for their purity using LC-MS/MS
and were found to have > 98% purity. Ovine COX-1 (Cat. No. 60100) and human COX-2 (Cat. No.
60122) were purchased from Cayman chemicals. All other chemicals were of high quality and used without further purification.
[00259] High-throughput Screen (HTS) [002601 HTS Materials. Dimethyl sulfoxide (DMS()) ACS grade was from Fisher, while ferrous ammonium sulfate, Xylenol Orange (XO), sulfuric acid, and Triton X-100 were obtained from Sigma-Aldrich.
[00261] HTS Compound library. A 74,290 compound library was screened in 7 to concentrations ranging from 0.7 nM to 57 gM. The library included 61,548 diverse small drug-like molecules that are part of the NIH Small Molecule Repository. A
collection of 1,372 compounds from the Centers of Methodology and Library Development at Boston University (BUCMLD) and University of Pittsburgh (UPCMLD) were added to the library.
Several combinatorial libraries from Pharmacopeia, Inc. totaled 2,419 compounds. An additional 1,963 compounds from the NCI Diversity Set were included. Lastly, 6,925 compounds with known pharmacological activity were added to provide a large and diverse screening collection.
[002621 HTS Protocol and analysis. All screening operations were performed on a fully integrated robotic system (Kalypsys Inc, San Diego, CA) as before (Kenyon et al., J. Med.

Chem., 54, 5485-5497). Three pL of enzyme (40 nM 15-hL0-1, final concentration) was dispensed into 1536-well Greiner black clear-bottom assay plate. Compounds and controls (23 nL) were transferred via Kalypsys PinTool equipped with 1536-pin array. The plate was incubated for 15 min at room temperature, and then a I pi, aliquot of substrate solution (50 p.M
arachidonic acid final concentration) was added to start the reaction. The reaction was stopped after 6.5 min by the addition of 4 AL FeX0 solution (final concentrations of 200 pM Xylenol Orange (XO) and 300 AM ferrous ammonium sulfate in 50 mM sulfuric acid). After a short spin (1000 rpm, 15 sec), the assay plate was incubated at room. temperature for 30 minutes. The absorbances at 405 and 573 nm were recorded using ViewLux high throughput CCD
im.ager (Perkin-Elmer, Waltham, MA) using standard absorbance protocol settings.
During dispense, enzyme and substrate bottles were kept submerged into +4 C recirculating chiller bath to minimize degradation. Plates containing DAS only (instead of compound solutions) were included approximately every 50 plates throughout the screen to monitor any systematic trend in the assay signal associated with reagent dispenser variation or decrease in enzyme specific activity.
1002631 Data was analyzed in a similar method as described before (Kenyon et al., J. Med.
Chem., 54, 5485-5497). Briefly, assay plate-based raw data was normalized to controls and plate-based data corrections were applied to filter out background noise. All concentration response curves (CRCs) were fitted using in-house developed software (http://ncgc.nih.gov/pub/openhts/).
Curves were categorized into four classes: complete response curves (Class 1), partial curves (Class 2), single point actives (Class 3) and inactives (Class 4). Compounds with the highest quality, Class 1 and Class 2 curves, were prioritized for follow-up.
1002641 Overexpression and Purification of Lipoxygenases. Different lipoxygenases such as, human reticulocyte 15-lipoxygenase-1 (12/15-LOX), human epithelial 15-lipoxygenase-2 (15-LOX-2), human platelet 12-lipoxygenase (12-LOX) were expressed as N-terminal His6-tagged proteins and were purified via immobilized metal affinity chromatography (IMAC) using Ni-NTA resins for 12/15-LOX and 15-LOX-2, whereas Ni-IDA resin for 12-LOX.7576 The protein purity was evaluated by SDS-PAGE analysis and was found to be greater than 90%. Human 5-lipoxygenase (5-LOX) was expressed as a non-tagged protein and used as a crude ammonium sulfate protein fraction, as published previously.77 [002651 Lipoxygenase UV-Vis Assay. The inhibitor compounds were screened initially using one concentration point on a Perkin-Elmer Lambda 40 UV-Vis spectrometer.
The percent inhibition was determined by comparing the enzyme rates of the control (DNB
solvent) and the inhibitor sample by following the formation of the conjugated diene product at 234 nrn (t: =
25,000 Mc m). The reactions were initiated by adding either of 40 nM 12-LOX, 40 nIVI
12/15-LOX, 0.5 AM 15-LOX-2 or. 200 nM 5-LOX (ammonium sulfate suspension) to a cuvette with a 2mL reaction buffer constantly stirred using a magnetic stir bar at room temperature (22"
C). It should be noted that LOX isozymes are often expressed in the inactive demetallated form, so it is best to utilize activity to determine the optimal LOX concentration for the assay (optimal rate of approximately 0.001 abs/see at 10 p.M AA). Reaction buffers used for various lipoxygenase were as follows- 25 rriM HEPES (pH 7.3), 0.3 mM CaCl2, 0.1 mM
EDTA, 0.2 mM
ATP, 0.01% Triton X-100, 10 iLtM AA for the crude, ammonium sulfate precipitated 5-LOX; 25 rriM Hepes (pH 8), 0.01% Triton X-100, 10 p.M AA for 12-LOX and 25 mM Hepes buffer (pH
7.5), 0.01% Triton X-100, 10 p.M AA for 12/15-LOX and 15-LOX-2. The substrate concentration was quantitatively determined by allowing the enzymatic reaction to go to completion in the presence of 15-LOX-2. For the inhibitors that showed more than 50%
inhibition at the one point screens (25 M inhibitor), IC50 values were obtained by determining the enzymatic rate at various inhibitor concentrations and plotted against inhibitor concentration (Approximate range: 0.1 to 25 p.M inhibitor), followed by a hyperbolic saturation curve fit (assuming total enzyme concentration [E] Kiam, so IC50 ¨ Kn. It should be noted that all of the potent inhibitors displayed greater than 80% maximal inhibition, unless stated in the tables.
Inhibitors were stored at -20 C in DMSO.
[002661 Pseudoperoxidase Assay. The pseudo-peroxidase activity rates were determined with BWb70c as the positive control, 13-(S)-HPODE as the oxidizing product and 12/15-LOX on a Perkin-Elmer Lambda 40 UV-Vis spectrometer, as described previously.78 Briefly, activity was determined by monitoring the decrease at 234 nm (product degradation) in buffer (50 mM
Sodium Phosphate (pH 7.4), 0.3 mM CaCl2, 0.1 mM EDTA, 0.01% Triton X100, and 20 pM 13-(S)-HPODE). 12/15-LOX was added to buffer (22 C) and the reaction initiated by addition of 20 p.M inhibitor (1:1 ratio to product). The percent consumption of 13-(S)-HPODE
was recorded, with individual controls being conducted with inhibitor alone with product and enzyme alone with product.

[002671 Steady-State Inhibition Kinetics. The steady-state kinetics experiments were performed with the parent analogue, compound 1 (ML351) to determine the mode of inhibition, as described before.56' 79 The inhibitor concentrations ranging from 0, 0.05, 2 and 5 tiM were used. Reactions were initiated by adding approximately 40-60 nM 12/15-LOX to a constantly stirring 2 mL cuvefte containing 1 ¨20 jaM AA in 25 mM HEPES buffer (pH 7.5), in the presence of 0.01% Triton X-100. Lipoxygenase rates were determined by monitoring the formation of the conjugated product, 15-HpETE, at 234 nm (c = 25 000 M-1 cm-1) with a Perkin-Elmer Lambda 40 IN-Vis spectrometer. The substrate concentration was quantitatively determined by allowing the enzymatic reaction to proceed to completion using 15-LOX-2.
Kinetic data were obtained by recording initial enzymatic rates, at varied substrate and inhibitor concentrations, and subsequently fitted to the Henri-Michaelis-Menten equation, using KaleidaGraph (Synergy) to determine the microscopic rate constants, Vn.õ
(pmollmin/mg) and V,õõõ/Km (pmollminlm.g/iiM). These rate constants were subsequently replotted with 1/V., and KmN. versus inhibitor concentration, yielding and K, respectively.
[002681 Cyclooxygenase Assay. About 2-5 jig of either COX-1 or COX-2 were added to buffer containing 0.1 M Tris-HCl buffer (pH 8.0), 5 mM EDTA, 2 mM phenol and 1 p.M
hematin at 37 C. The inhibitors were added to the reaction cell, followed by an incubation of 5 minutes with either of the COX. enzymes. The reaction was then initiated by adding 1001.IM AA
in the reaction cell. Data was collected using a Hansatech DWI oxygen electrode and the consumption of oxygen was recorded. Indomethacin and the solvent DMSO, were used as positive and negative controls, respectively and the percent inhibition of the enzyme was calculated by comparing the rates from samples and the controls.
(00269) HT22 Cell Culture Assay. Glutathione depletion was induced in HT22 cells by glutamate treatment, and LDH release into the medium was measured to detect cell death as described.38 Briefly, HT22 cells were cultured in DMEM containing 10% fetal bovine serum and penicillin / streptomycin (all media from Invitrogen). For viability experiments, cells were seeded at 1 x 104 cells/well in 96-well plates (Corning) and treated 18h later, when the cells were approximately 50-70% confluent. Treatment consisted of exchanging the medium to 100 pi fresh culturing medium and adding 5 mM glutamate (stock solution 0.5 M in PBS) in the presence or absence of DMSO (maximum 0.1% final concentration) as control or the indicated concentrations of!. Lactate dehydrogenase (LDH) content was determined separately for the cell extracts and corresponding media using a Cytotoxicity Detection Kit (Roche), and the percentage of LDH released to the medium calculated after subtracting the corresponding background value.
To determine levels of the 12/15-LOX metabolite, 12-HETE, HT22 cells were cultured in 75 cm2 flasks in DMEM medium without phenol red, supplemented with 5% FBS, and treated the cells the next day when cells were 50-70% confluent. 24 hours later, the eicosan.oid-containing fraction was isolated via Sep-Pak C-18 column, and 12-HETE was detected with a ELISA kit (En.zo Life Sciences), used according to the manufacturer's instruction.. Three independent experiments were evaluated. For the MS samples, the above procedure was modified slightly. The eicosanoids containing fractions were transferred to scintillation vials followed by addition of perdeuterated. 13-HODE (13-d31-HODE), as an internal control for extraction and 1.5% glacial acetic acid for protein precipitation. The samples were extracted with methylene chloride, reduced with trimethylphosphite and evaporated to dryness.
The dry samples were then re-constituted in methanol and an internal control 12-deuterated (d8)- HETE (12-d8-BETE), was added to each sample for detector response variation and it was assumed that the change in detector response for 12-HETE and 12- d8-FiETE would be similar. The samples were run on and analyzed by Finnigan LTQ liquid chromatography--tandem mass spectrometry (LC---MS/MS) system. A Thermo Electron Corp. Aquasil (3 gm, 100 mm x 2.1 mm) C-column was used to detect the ITETEs with an elution protocol consisting of 0.2 mL /min flow rate and a linear gradient from 54.9% ACN, 45% H.20, and 0.1% THF to 69.9% AN, 30% H20, and 0.1% THF. The electrospray voltage was set to 5.0 kV and a global acquisition MS mode was used. The MS-MS scan was performed for the five most abundant precursor ions. The Collision Induced Dissociation (CID) was used for MS-MS with collision energy of 35 eV. The corresponding 12-HETE, 12-d8-HETE and 13-d31-HODE compounds were detected using selective ion monitoring analysis (Luiz ¨318.7 to 319.7, 326.8 to 327.7 and 325.8 to 326.8 respectively') in negative ion mode and then identified by fragmentation pattern (12-HETE, parent ion at m/z 319 and fragments at m/z 179 and 163; 12-d8-HETE, parent ion at miz 327 and fragments at m/z 184 and 13-d31-110DE, parent ion m/z 326 and fragments at mlz 213) from MS-MS. The peak area of 12-d8-HETE for each sample was normalized to the area of
13-d3i-HODE.
The peak intensities of 12-HETEs were normalized to the corrected 12-d8-HETE
intensities. The amount of 12-HETE in samples was estimated from the pure 12-(S)-HETE standard curve.

[002701 Distal MCAO Model of Permanent Focal Ischemia in Mice. To study 1 in a model of distal middle cerebral artery occlusion (distal MCAO), 74 C57B16,1 mice were treated with ferric chloride (FeCI3) to cause occlusion of the distal middle cerebral artery. Mice were kept under anesthesia with 1.5 % isoflurane in a nitrous oxide/oxygen mixture via facemask. The body temperature was monitored by a rectal probe and maintained at 37 0.3 C
by a homoeotherm.ic blanket control unit. Briefly, mice were placed in a stereotaxic frame, the scalp was opened and right temporal muscle was dissected. The area between zygom.atic arch and squamous bone was thinned by a high-speed drill and cooled with saline. The trace of MCA was visualized and thin bony film was lifted up by forceps. After that a laser-Doppler flowmetry probe was placed 2 mm posterior, 6 mm. lateral to the bregm.a to monitor the regional cerebral blood flow (rCBF). After obtaining a stable epoch of the pre-ischemic rCBF, a piece of 10%
FeC13 saturated filter paper was placed over the intact dura mater along the trace of MCA and the 1C,B17 was continuously monitored during the next 3 hours. After 2 hours of ischemia, either 50 mg/kg I, or DMSO vehicle was injected intraperitoneally. Following sacrifice at 24 hours, brains were sectioned into imm slices, and infarct sizes were determined by staining with 2,3,5-triphenyltetrazolium chloride (ITC), using the indirect method (infarct volume = contralateral volume minus uninfarcted ipsilateral volume). Both the surgeon carrying out the procedure, as well as the researcher determining infarct volumes were blinded as to which treatment the mice received.
[00271.) Abbreviations. LOX, lipoxygenase; sLOX-1, soybean lipoxygenase-1;
12/15-LOX, human reticulocyte 15-lipoxygenase-1; 15-LOX-1, human reticulocyte 15-lipoxygenase-1; 15-LOX-2, human epithelial 15-lipoxygenase-2; 12-LOX, human platelet 12-lipoxygenase; rabbit 12/15-LOX, rabbit reticulocyte 12/15-lipoxygenase; mouse 12/15-LOX, mouse reticulocyte 12/15-lipoxygenase; COX, Cyclooxygenase; NDGA, nordihydroguaiaretic acid; AA, arachidonic acid; 15-HpETE, 15-(S)-hydroperoxyeicosatetraenoic acid; 12-HpETE, 12-(S)-hydroperoxyeicosatetraenoic acid; LA, linoleic acid; 13-(S)-HpODE, 13-(S)-hydroperoxyoctadecadienoic acid; 12-HETE, 12-(S)-hydroxyeicosatetraenoic acid;
15-HETE, 15-(S)-hydroxyeicosatetraenoic acid; V.; maximal velocity (mol/min); KM, Henri-Michaelis-Menten Constant (1.tM); [E], total active enzyme concentration; IC50, inhibitor constant at 50%
inhibition; KiaPP, apparent inhibition constant when [E] >> KiaPP; X0, xylenol orange; FITS, high-throughput screening; MLSMR, Molecular Libraries Small Molecule Repository;
MLPCN, Molecular Libraries Probe Centers Network; qHTS, quantitative high-throughput screening;
CRC, concentration response curve; EDC, 1-Ethyl-3-(3-dimethylaminopropyl)carbodiimide hydrochloride; BTAC, N-benzyl-triethylanamonium chloride; BWB70C, N-[343-(-Fluorophenoxy)pheny1]-1-methy1-2-propenyll-N-hydroxyurea; LDA, lithium diisopropylamide.
[002721 References for Example 1:
1. Solomon, E. I.; Zhou, J.; Neese, F.; Pavel, E. G. New Insights from.
Spectroscopy into the Structure/Function Relationships of Lipoxygenases. Chem. Biol. 1997, 4, 795-808.
2. Brash, A.. R. Lipoxygenases: Occurrence, Functions, Catalysis and Acquisition of Substrate. J. Biol. Chem. 1999, 274, 23679-23682.
3. Ivanov, I.; Heydeck, D.; Hoflieinz, K..; Roffeis, J.; O'Donnell, V. B.;
Kuhn, H.; Walther, M. Molecular Enzymology of Lipoxygenases. Arch. Biochem. .Biophys. 2010, 503, 161-174.
4. Schwa, K.; Belkner, S.; Ursini, F.; Schevve, T.; Kuhn, H. The Selenoenzyme Phospholipid Hydroperoxide Glutathione Perox.idase Controls the Activity of the 15-Lipoxygenase with Complex Substrates and Preserves the Specificity of the Oxygenation Products../. Biol. Chem. 1996, 271, 4653-4658.
5. Tong, W. G.; Ding, X. Z.; Hennig, R.; Witt, R. C.; Standop, j.; Pour, P.
M.; Adrian, T. E.
Leukotriene B4 Receptor Antagonist Ly293111. Inhibits Proliferation and induces Apoptosis in Human Pancreatic Cancer Cells. Clin. Can. Res. 2002, 8, 3232-3242.
6. Wenzel, S. E.; K.amada, A. K. Zileuton: The First 5-Lipoxygenase Inhibitor for the Treatment of Asthma. Ann. Pharmacother. 1996, 30, 858-864.
7. O'Byme, P. M.; Israel, E.; Drazen, J. M. .Antileukotrienes in the Treatment of Asthma.
Annals Int. Med. 1997, 127, 472-480.
8. Berger, W.; De Chandt, M. T.; Cairns, C. B. Zikuton: Clinical Implications of 5-Lipoxygenase Inhibition in Severe Airway Disease. Int. J. Clin. Praa. 2007, 61, 663-6676.
9. Muller-Peddinghaus, R. Potential Anti-Inflammatory Effects of 5-Lipoxygenase Inhibition--Exemplified by the Leukotriene Synthesis Inhibitor Bay X 1005. J.
Physiol.
Pharmaco.11997, 48, 529-536.
10. Klickstein, L. B.; Shapleigh, C.; Goetzl, E. J. Lipoxygenation of Arachidonic Acid as a Source of Polymorphonuclear Leukocyte Chemotactic Factors in Synovial Fluid and Tissue in Rheumatoid Arthritis and Spondyloarthritis. J. Clin. Investig. 1980, 66, 1166-1170.

11. Weinblatt, M. E.; Kremer, J. M.; Coblyn, J. S.; Helfgott, S.; Maier, A.
L.; Petrillo, G.;
Henson, B.; Rubin, P.; Sperling, R. Zileuton, a 5-Lipoxygenase Inhibitor in Rheumatoid Arthritis. J. RheumatoL 1992, 19, 1537-1541.
12. Spanbroek, R.; Grabner, R.; Lotzer, K.; Hildner, M.; Urbach, A.;
Ruhling, K.; Moos, M.
P.; Kaiser, B.; Cohnert, T. U.; Wahlers, T.; Zieske, A.; Plenz, G.; Robenek, H.; Salbach, P.;
Kuhn, H.; R.admark, 0.; Samuelsson, B.; Habenicht, .A. J. Expanding Expression of the 5-Lipoxygenase Pathway within the Arterial Wall During Human Atherogenesis.
.P.N.A.S. 2003, 100, 1238-1243.
13. Funk, C. D. Leukotriene Modifiers as Potential Therapeutics for Cardiovascular Disease.
Nat. Rev. Drug Discovery 2005, 4, 664-6672.
14. Lotzer, K.; Funk, C. D.; HabenichtõA.. J. The 5-Lipoxygenase Pathway in Arterial Wall Biology and Atherosclerosis. Biochim. Biophys. Ada. 2005, /736, 30-37.
15. Bleich, D.; Chen, S.; Gu, J. L.; Nadler, J. L. The Role of 12-Lipoxygenase in Pancreatic -Cells (Review). Int. J. MO/. Med. 1998, 1, 265-272.
16. Hedrick, C. C.; Kim, M. D.; Natarajan, R. D.; Nadler, J. L. 12-Lipoxygenase Products Increase Monocyte:Endothelial Interactions. Adv. Exp. Med Biol. 1999, 469, 455-460.
17. Thomas, C. P.; Morgan, L. T.; Maskrey, B. H.; Murphy, R. C.; Kuhn, H.;
Hazen, S. L.;
Goodall, A. H.; Hamali, H. A.; Collins, P. W.; O'Donnell, V. B. Phospholipid-Esterified Eicosanoids Are Generated in Agonist-Activated Human Platelets and Enhance Tissue Factor-Dependent Thrombin Generation../. Biol. Chem. 2010, 285, 6891-6903.
18. Hussain, H.; Shomick, L. P.; Shannon, V. R.; Wilson, J. D.; Funk, C.
D.; Pentland, A. P.;
Holtzman, M. J. Epidermis Contains Platlet-Type 12-Lipoxygenase That Is Overexpressed in Germinal Layer Keratinocytes in Psoriasis. Am. Physiol. 1994, 266, C243-C253.
19. Connolly, J. M.; Rose, D. P. Enhanced Angiogenesis and Growth of 12-Lipoxygenase Gene-Transfected Mcf-7 Human Breast Cancer Cells in Athymic Nude Mice. Cancer Lett. 1998, 132,107-112.
20. Ding, X. Z.; :Iversen, P.; Cluck, M. W.; Knezetic, J. A.; Adrian, T. E.
Lipoxygenase inhibitors Abolish Proliferation of Human Pancreatic Cancer Cells.
Blochem.Blophys. Res.
Comm. 1999, 261, 218-223.
21. Haeggstrom, J.; Funk, C. D. Lipoxygenase and Leukotriene Pathways:
Biochemistry, Biology, and Roles in Disease. Chem. Rev. 2011, 111, 5866-5898.
22. Kuhn, H.; Walther, M.; Kuban, R. J. Mammalian Arachidonate 15-Lipoxygenases:
Structure, Function, and Biological Implications. Prostag. Lipid M. 2002, 68-69, 263-290.
23. Weaver, J. R.; Holman, T. R.; Imai, Y.; Jadhav, A.; Kenyon, V.;
Maloney, D. J.; Nadler, J. L.; Rai, G.; Simeonov, A.; Taylor-Fishwick, D. A. Integration of Pro-Inflammatory Cytokines, 12-Lipoxygenase and Nox-1 in Pancreatic Islet Beta Cell Dysfunction. Mol.
Cell. Endo. 2012, 358, 88-95.
24. Stavniichuk, R..; Drel, V. R..; Shevalye, H.; Vareniuk, I.; Stevens, M.
J.; Nadler, J. L.;
Obrosova, I. G. Role of 12/15-Lipoxygenase in Nitrosative Stress and Peripheral Prediabetic and Diabetic Neuropathies. Free Rad. Bio. & Med. 2010, 49, 1036-1045.
25. Obrosova, I. G.; Stavniichuk., R.; Drel, V. R.; Shevalye, H.; Vareniuk, I.; Nadler, J. L.;
Schmidt, R.. E. Different Roles of 12/15-Lipoxygenase in Diabetic Large and Small Fiber Peripheral and Autonomic Neuropathies. Am. J. Path. 2010, 177, 1436-1447.
26. Dobrian, A. D.; Lieb, D. C.; Ma, Q.; Lindsay, J. W.; Cole, B...; Ma, K.; Chakrabard, S.
K.; Kuhn, N. S.; Wohlgemuth, S. D.; Fontana, M.; Nadler, J. L. Differential Expression and Localization of 12/15 Lipoxygenases in Adipose Tissue in Human Obese Subjects.
Biochem.
Biophys. Res. Comm. 2010, 403, 485-490.
27. Chakrabarti, S. K.; Wen, Y.; Dobrian, A. D.; Cole, B. K.; Ma, Q.; Pei, 171.; Williams, M.
D.; Bevard, M. H.; Vandenhoff, G. E.; Keller, S. R.; Gu, J.; Nadler, S. L.
Evidence for Activation of Inflammatory Lipoxygenase Pathways in Visceral Adipose Tissue of Obese Zucker Rats. Am.
J. Physiol. Endo. Metab. 2010, 300, E175-E187.
28. Nunemaker, C. S.; Chen, M.; Pei, H.; Kimble, S. D.; Keller, S. R.;
Carter, J. D.; Yang, Z.;
Smith, K. M.; Wu, R.; Bevard, M. H.; Garmey, J. C.; Nadler, J. L. 12-Lipoxygenase-Knockout Mice Are Resistant to Inflammatory Effects of Obesity Induced by Western Diet.
Am. J. Physiol.
Endo. Metab. 2008, 295, E1065-E1075.
29. Succol, F.; Pratico, D. A Role for 12/15 Lipoxygenase in the Amyloid Beta Precursor Protein Metabolism. J. Neurochem. 2007, 103, 380-387.
30. Yao, Y.; Clark, C. M.; Trojanowski, S. Q.; Lee, V. M.; Pratico, D.
Elevation of 12/15 Lipoxygenase Products in Ad and Mild Cognitive impairment. Ann. Neurol. 2005, 58, 623-626.
31. Pratico, D.; Zhukareva, V.; Yao, Y.; Uryu, K.; Funk, C. D.; Lawson, J.
A.; Trojanowski, J. Q.; Lee, V. M. 12/15-Lipoxygenase Is Increased in Alzheimer's Disease:
Possible Involvement in Brain Oxidative Stress. Am. J. Pathol. 2004, 164, 1655-1662.
32. Haynes, R. L.; van Leyen, K. 12/15-Lipoxygenase Expression Is Increased in Oligodendrocytes and Microglia of Periventricular Leukomalacia. DeveL Neuro.
2013, 35, 140-154.
33. O'Flaherty, J. T.; Wooten, R. E.; Samuel, M. P.; Thomas, M. J.; Levine, E. A.; Case, L.
D.; Akman, S. A.; Edwards, I. J. Fatty Acid Metabolites in Rapidly Proliferating Breast Cancer.
PloS one 2013,8, e63076.
34. Yigitkanli, K..; Pekcec, A.; Karatas, H.; Pallast, S.; Mandeville, E.;
Joshi, N.; Smimova, N.; Gazaryan, I.; Ratan, R. R.; Witztum, J. L.; Montaner, J.; Holman, T. R.;
Lo, E. H.; van Leyen, K. Inhibition of 12/15-Lipoxygenase as Therapeutic Strategy to Treat Stroke. Ann.
NeuroL 2013, 73, 129-135.
35. van Leven, K. Lipoxygenase: An Emerging Target for Stroke Therapy. CNS
NeuroL
.Disord. Drug Targets 2013, 12, 191-199.
36. Lo, E. H.; Dalkara, T.; Moskowitz, M. A. Mechanisms, Challenges and Opportunities in Stroke. Nat. Rev. Neurosci. 2003, 4, 399-415.
37. Donnan, G. A.; Fisher, M.; Macleod, M.; Davis, S. M. Stroke. Lancet.
2008, 37/, 1612-1623.
38. Li, Y.; Maher, P.; Schubert, D. A Role for 12-Lipoxygenase in Nerve Cell Death Caused by Glutathione Depletion. Neuron 1997, 19, 453-463.
39. Lovat, P. E.; 01.iverio, S.; Ranalli, M.; Corazzari, M.; Rodolfo, C.;
Bemassola, F.;
Aughton, K.; Maccarrone, M.; Hewson, Q. D.; Pearson, A. D.; Melino, G.;
Piacentini, M.;
Redfern, C. P. Gadd153 and 12-Lipoxygenase Mediate Fenretinide-Induced Apoptosis of Neuroblastoma. Cancer Res. 2002, 62, 5158-5167.
40. Canals, S.; Casarejos, M. j.; de Bernardo, S.; Rodriguez-Martin, E.;
Mena, M. A. Nitric Oxide Triggers the Toxicity Due to Glutathione Depletion in Midbrain Cultures through 12-Lipoxygenase. .1. Biol. Chem. 2003, 278, 21542-21549.
41. Khanna, S.; Roy, S.; Ryu, H.; Bahadduri, P.; Swaan, P. W.; Ratan, R.
R.; Sen, C. K.
Molecular Basis of Vitamin E Action: Tocotrienol Modulates 12-Lipoxygenase, a Key Mediator of Glutamate-Induced Neurodegeneration. J. Biol. Chem. 2003, 278, 43508-43515.
42. Khanna, S.; Roy, S.; Slivka, A.; Craft, T. K.; Chaki, S.; Rink, C.;
Notestine, M. A.;
DeVries, A. C.; Parinandi, N. L.; Sen, C. K. Neuroprotective Properties of the Natural Vitamin E
Alpha-Tocotrienol. Stroke 2005, 36, 2258-2264.
43. van Leyen, K.; Kim, H. Y.; Lee, S. R.; Jin, G.; Arai, K.; Lo, E. H.
Baicalein and 12/15-Lipoxygenase in the Ischemic Brain. Stroke 2006, 37, 3014-3018.
44. van Leyen, K.; Lee, S. R.; Siddig, A.; Ratan, R. R.; Lo, E. H. 12/15-Lipoxygenase and the Proteasome as Mediators of Neuronal Oxidative Stress and Stroke. Program No 1356, 2004 Abstract Viewer/Itinerary Planner. Washington, DC: S'ocietylbr Neuroscience 2004.
45. jin, G.; Arai, K.; Murata, Y.; Wang, S.; Stins, M. F.; Lo, E. H.; van Leyen, K. Protecting against Cerebrovascular Injury: Contributions of 12/15-Lipoxygenase to Edema Formation Following Transient Focal Ischemia. Stroke 2008, 39, 2538-2543.
46. Pallast, S.; .Arai, K.; Wang, X.; Lo, E. H.; van Leyen, K. 12/15-Lipoxygenase Targets Neuronal Mitochondria under Oxidative Stress. J. Newochem. 2009, 1H , 882-889.
47. Pallast, S.; Arai, K.; Pekcec, A.; Yitkartli, K.; Yu, Z.; Wang, X.; Lo, E. H.; van Leyen, K. Increased Nuclear Apoptosis-inducing Factor after Transient Focal Ischemia:

Lipoxygenase-Dependent Organelle Damage Pathway. J. Cereb. Blood Flow Metab.
2010, 30, 1157-1167.
48. Seiler, A.; Schneider, M.; Forster, H.; Roth, S.; Wirth, E. K.;
C,ulmsee, C.; Plesnila, N.;
Kremmer, E.; Radmark, 0.; Wurst, W.; Bornkamm, G. W.; Schweizer, U.; Conrad, M.
Glutathione Peroxidase 4 Senses and Translates Oxidative Stress into 12/15-Lipoxygenase Dependent- and Aif-Mediated Cell Death. Cell Metab. 2008, 8, 237-248.
49. Kenyon, V.; Chorny, I.; Carvajal, W. J.; Holman, T. R.; Jacobson, M. P.
Novel Human Lipoxygenase Inhibitors Discovered Using Virtual Screening with Homology Models. f. Med.
Chem. 2006, 49, 1356-1363.
50. Carroll, j.; jonsson, E. N.; Ebel, R..; Hartman, M. S.; Holman, T. R.;
Crews, P. Probing Sponge-Derived Terpenoids for Human 15-Lipoxygenase Inhibitors. J. Org. Chem.
2001, 66, 6847-6851.
51. Whitman, S.; Gezginci, M.; Titnmermann, B. N.; Holman, T. R. Structure-Activity Relationship Studies of Nordihydroguaiaretic Acid Inhibitors toward Soybean, 12-Human, and 15-Human Lipoxygenase. J. Med. Chem. 2002, 45, 2659-2661.
52. Amagata, T.; Whitman, S.; Johnson, T. A.; Stessman, C. C.; Loo, C. P.;
Lobkovsky, E.;
Clardy, J.; Crews, P.; Holman, T. R. Exploring Sponge-Derived Terpenoids for Their Potency and Selectivity against 12-Human, 15-Human, and 15-Soybean Lipoxygenases. J.
Nat. Prod.
2003, 66, 230-235.
53. Cichewicz, R. H.; Kenyon, V. A.; Whitman, S.; Morales, N. M.; Arguello, J. F.; Holman, T. R.; Crews, P. Redox Inactivation of Human 15-Lipoxygenase by Marine-Derived Meroditerpenes and Synthetic Chromanes: Archetypes for a Unique Class of Selective and Recyclable Inhibitors. J. Am. Chem. Soc. 2004, 126, 14910-14920.
54. Vasquez-Martinez, Y.; Obri, R. V.; Kenyon, V.; Holman, T. R.; Sepulveda-Boza, S.
Structure-Activity Relationship Studies of Flavonoids as Potent inhibitors of Human Platelet 12-Hlo, Reticulocyte 15-Hlo-1, and Prostate Epithelial 15-Hlo-2. Bio. AfecL Chem.
2007, 15, 7408-7425.
55. Deschamps, J. D.; Kenyon, V. A.; Holman, T. R. Baicalein Is a Potent in Vitro Inhibitor against Both Reticulocyte 15-Human and Platelet 12-Human Lipox.ygenases.
Bloorg. Med.
Chem. 2006, 14, 4295-4301.
56. Rai, G.; Kenyon, V.; Jadhav, A.; Schultz, L.; Armstrong, M.; Jameson, J. B.; Hoobler, E.;
Leister, W.; Simeonov, A.; Holman, T. R.; Maloney, D. J. Discovery of Potent and Selective Inhibitors of Human Reticulocyte 15-Lipoxygenase-1.../. Med. Chem. 2010, 53, 7392-7404.
57. Ngu, K.; Weinstein, D. S.; Liu, W.; Langevine, C.; Combs, D. W.;
Zhuang, S.; Chen, X.;
Madsen, C. S.; Harper, T. W.; Ahm.ad, S.; R.obl, J. A. Pyrazole-Based Sulfonamide and Sulfamides as Potent Inhibitors of Mammalian 15-Lipoxygena.se. Bio. Med. Chem.
Let. 2011, 21, 4141-4145.
58. Weinstein, D. S.; Liu, W.; (hi, Z.; Langevine, C.; Ngu, K.; Fadnis, L.;
Combs, D. W.;
Sitkoff, D.; Ahmad, S.; Zhuang, S.; Chen, X.; Wang, F. L.; Loughney, D. A.;
Atwal, K. S.;
Zahler, R.; Macor, J. E.; Madsen, C. S.; Murugesan, N. Tryptamine and Homotryptamine-Based Sulfonamides as Potent and Selective inhibitors of 15-Lipoxygenase. Bio. Med.
Chem. Let. 2005, 15, 1435-1440.
59. Weinstein, D. S.; Liu, W.; Ngu, K.; Langevine, C.; Combs, D. W.;
Zhuang, S.; Chen, C.;
Madsen, C. S.; Harper, T. W.; Rob!, J. A. Discovery of Selective Imidazole-Based Inhibitors of Mammalian 15-Lipoxygenase: Highly Potent against Human Enzyme within a Cellular Environment. Bio. Med. Chem. Let. 2007, 17, 5115-5120.
60. Malterud, K. E.; Rydland, K. M. Inhibitors of 15-Lipoxygenase from Orange Peel. J. Ag.
Food Chem. 2000, 48, 5576-5580.
61. Sailer, E. R.; Schweizer, S.; Boden, S. E.; Ammon, H. P. T.; Safayhi, H. Characterization of an Acety1-11-Keto-B-Boswellic Acid and Arachidonate-Binding Regulatory Site of 5-Lipoxygenase Using Phoroaffinity Labeling. Eur. J. Biochem. 1998, 256, 364-368.
62. Sendobry, S. M.; CorniceIli, J. A.; Welch, K.; Bocan, T.; Tait, B.;
Trivedi, B. K.; Colbry, N.; Dyer, R. D.; Feinmark, S. J.; Daugherty, A. Attenuation of Diet-Induced Atherosclerosis in Rabbits with a Highly Selective 15-Lipoxygenase Inhibitor Lacking Significant Antioxidant Properties. British J. pharniacol. 1997, 120, 1199-1206.
63. Diana, G. D.; Treasurywala, A.. M.; Bailey, T. R..; Oglesby, R.. C.;
Pevear, D. C.; Dutko, F. J. A Model for Compounds Active against Human Rhinovirus-14 Based on X-Ray Crystallography Data. J. Med. Chem. 1990, 33, 1306-1311.
64. Xia, Q.; Ganem, B. Metal-Mediated Variants of the Passerini Reaction: A
New Synthesis of 4-Cyanooxazol es. Synthesis 2002, 14, 1969-1972.
65. Dewar, M. J. S.; Turchi, I. J. Cornforth Rearrangement. J. Am. Chem.
Soc. 1974,96, 6148-6152.
66. Boros, E. E.; johns, B. A.; Garvey, E. P.; Koble, C. S.; Miller, W. H.
Synthesis and Hiv-Integrase Strand Transfer Inhibition Activity of 7-Hydroxy[1,3]Thiazolo[5,4-B]Pyridin-5(4h)-Ones. .Bio. Med. Chem. Let. 2006, 16, 5668-5672.
67. Ratan, R. R.; Murphy, T. H.; Baraban, J. M. Oxidative Stress Induces Apoptosis in Embryonic Cortical Neurons. J. Neurochem. 1994, 62, 376-379.
68. Ratan, R. R.; Ryu, H.; Lee, J.; Mwidau, A.; Neve, R. L. In Vitro Model of Oxidative Stress in Cortical Neurons. Methods Enzymol. 2002, 352, 183-190.
69. Murphy, T. H.; Schnaar, R. L.; Coyle, J. T. Immature Cortical Neurons Are Uniquely Sensitive to Glutamate Toxicity by Inhibition of Cystine Uptake. FASEB J
1990,4, 1624-1633.
70. van Leyen, K.; Siddiq, A.; Ratan, R. R.; La, E. H. Proteasome inhibition Protects Ilt22 Neuronal Cells from Oxidative Glutamate Toxicity. .1. Neurochem. 2005, 92, 824-830.
71. Zhang, J. H.; Chung, T. D.; Oldenburg, K. R. A Simple Statistical Parameter for Use in Evaluation and Validation of High Throughput Screening Assays. ,J. BiomoL
Screen 1999,4, 67-73.
72. Mogul, R.; Johansen, E.; Holman, T. R. ley]. Sulfate Reveals Allosteric Inhibition of Soybean Lipoxygenase-1 and Human 15-Lipoxygenase. Biochemistry 2000, 39, 4801-4807.
73. Weeksler, A. T.; Kenyon, V.; Deschamps, J. D.; Holman, T. R. Substrate Specificity Changes for Human Reticulocyte and Epithelial 15-Lipoxygenases Reveal Allosteric Product Regulation. Biochemistry 2008, 47, 7364-75.
74. Karatas, H.; Erdener, S. E.; Gursoy-Ozdemir, Y.; Gurer, G.;
Soyletnezoglu, F.; Dunn, A.
K.; Dalkara, T. Thrombotic Distal Middle Cerebral Artery Occlusion Produced by Topical Fec1(3) Application: A Novel Model Suitable for intravital Microscopy and Throm.bolysis Studies. J. Cereb. Blood Flow Metab. 2011, 31, 1452-1460.
75. Ohri, R. V.; R.adosevich, .A. T.; Hrovat, K. J.; Musick C.; Huang, D.;
Holman, T. R.;
Toste, F. D. A R.e(V)-Catalyzed C-N Bond-Forming Route to Human Lipoxygenase Inhibitors.
Org. Let. 2005, 7, 2501-2504.
76. Chen, X. S.; Brash, A. R..; Funk, C. D. Purification and Characterization of Recombinant Histidine-Tagged Human Platelet 12-Lipoxygenase Expressed in a Baculovirusiinsect Cell System. Eur. J. Biochem. 1993, 214, 845-852.
77. Robinson, S. J.; Hoobler, E. K.; Riefler, M.; Loveridge, S. T.; Tenney, K..; Valeriote, F.
A.; Holman, T. R.; Crews, P. Using Enzyme Assays to Evaluate the Structure and Bioactivity of Sponge-Derived Meroterpenes../. Nat. Prod. 2009, 72, 1857-1863.
78. Hoobl.er, E. K.; Holz, C.; Holman, T. R. Pseudoperoxidase Investigations of Hydroperoxides and Inhibitors with Human Lipoxygenases. Bioorg. Med. Chem.
2013,21, 3894-3899.
79. Kenyon, V.; Rai, G.; Iadhav, A.; Schultz, L.; Armstrong, M.; Jameson, J. B., 2nd; Perry, S.; Josh, N.; Bougie, J. M.; Leister, W.; Taylor-Fishwick, D. A.; Nadler, J.
L.; Holinstat, M.;
Simeonov, A.; Maloney, D. J.; Holman, T. R. Discovery of Potent and Selective inhibitors of Human Platelet-Type 12- Lipoxygenase..1. Med. Chem. 2011,54, 5485-5497.
Example 2: Effect on Warfarin-associated hemorrhage following experimental stroke in mice [00273] The inventors have shown that ML351 can reduce infarct size in a mouse model of transient focal ischemia (see Example 1), indicating it is protective in cases of stroke. These studies arc expanded on by investigating possible benefits of ML351 in eases of stroke with hemorrhagic transformation, reflecting the clinical situation of patients who are on oral anticoagulants, specifically warfarin. Warfarin is typically given as a blood thinner to patients with atrial fibrillation who are at a 5-fold increased risk of ischernic stroke. Around 20% of all stroke patients over 80 years old have atrial fibrillation (Heart Disease ad Stroke Statistics - 2014 Update; Circulation (2014) 129(3):399-410. doi:
10.1161/01.cir.0000442015.53336.12).
[002741 Because these anticoagulants are seen as increasing the risk of bleeding (known as hemorrhagic transformation), this makes patients ineligible for treatment with tPA in case of stroke, because tPA similarly has an increased risk of hemorrhage as side effect. Patients on warfarin who experience a stroke, thus do not receive any form of drug treatment. To test the hypothesis that inhibition of 12/15-LOX may reduce hemorrhagic transformation, ML351 are tested in two different scenarios:
[002751 A) In warfarin-pretreated mice subjected to transient focal ischemia with 3h occlusion, 50 mg/kg ML351 was administered by i.p. injection 3h after onset of ischemia (FIG.
9A). This mimics the situation of a patient on warfarin who experiences a severe stroke with hemorrhagic transformation.
[002761 B) To investigate if administering tPA to patients with an ischemic stroke who are on warfarin can be made safe, warfarin-pretreated mice were subjected to 2b of transient focal ischemia, then treated with tPA 4h after onset of ischemia, and administered 50 mg/kg ML351 by i.p. injection concurrently with tPA. Again, vehicle-treated mice experienced massive hemorrhage, and again this was significantly reduced by ML351 (FIG. 9B).
[002771 These experiments show that patients who are on oral anticoagulants such as warfarin and experience a stroke, could now be treated either with ML351 as stand-alone treatment, or with ML351 as adjuvant to tPA. This widens the patient pool eligible for treatment following a stroke.

Claims (11)

What is claimed is:
1. Use of a compound for the preparation of a medicament for the treatment of a condition selected from stroke, periventricular leukomalacia and cardiac arrest with resuscitation, wherein the compound is of:
(i) Formula II:
or a pharmaceutically acceptable salt thereof, wherein:
X is 0; and R" is 2-naphthyl, 5-isoquinolinyl, 6-isoquinolinyl, 5-quinolinyl, 2,3-dichlorophenyl, 3,4-dichlorophenyl, 5-benzo[d][1,3]dioxolyl, 3-1H-indolyl, or chlorophenyl;
(ii) Formula III:
or a pharmaceutically acceptable salt thereof, wherein:
R12 is hydrogen; and R13 is ethyl, 1-propyl, 1-butyl, 1-pentyl, or allyl; or or a pharmaceutically acceptable salt thereof.
2. The use of claim 1, wherein the compound is 2-(2,3-dichloropheny1)-5-(methylamino)-1,3-oxazole-4-carbonitrile, 2-(3,4-dichloropheny1)-5-(methylamino)-1,3-oxazole-carbonitrile, or 5-(methylamino)-2-naphthalen-1-y1-1,3-thiazole-4-carbonitrile.
3. The use of claim 1, wherein the compound is 5-(ethylamino)-2-naphthalen-1-y1-1,3-oxazole-4-carbonitrile; 5-(propylamino)-2-naphthalen-1-y1-1,3-oxazole-4-carbonitrile; or 5-(butylamino)-2-naphthalen-1-y1-1,3-oxazole-4-carbonitrile.
4. The use of any one of claims 1-3, wherein the condition is stroke or periventricular leukomalacia.
5. The use of any one of claims 1-4, wherein the condition is stroke.
6. The use of any one of claims 1-5, wherein the subject is a mammal.
7. The use of any one of claims 1-6, wherein the subject is a human.
8. A compound selected from the group consisting of 2-(2,3-dichloropheny1)-(methylamino)-1,3-oxazole-4-carbonitrile; 2-(3,4-dichloropheny1)-5-(methylamino)-1,3-oxazole-4-carbonitrile; 5-(methylamino)-2-naphthalen-1-y1-1,3-thiazole-4-carbonitrile;
2-(6-isoquinoliny1)-5-(methylamino)-1,3-oxazole-4-carbonitrile; 5-(ethylamino)-naphthalen-1-y1-1,3-thiazole-4-carbonitrile; 5-(propylamino)-2-naphthalen-1-y1-1,3-thiazole-4-carbonitrile; 5-(butylamino)-2-naphthalen-1-y1-1,3-thiazole-4-carbonitrile; 5-(pentylamino)-2-naphthalen-1-y1-1,3-thiazole-4-carbonitrile; and 5-(allylamino)-2-naphthalen-1-y1-1,3-thiazole-4-carbonitrile.
9. The compound of claim 8, wherein the compound is 2-(2,3-dichloropheny1)-(methylamino)-1,3-oxazole-4-carbonitrile, 2-(3,4-dichloropheny1)-5-(methylamino)-1,3-oxazole-4-carbonitrile, or 5-(methylamino)-2-naphthalen-1-y1-1,3-thiazole-4-carbonitrile.
10. The compound of claim 8, wherein the compound is 5-(ethylamino)-2-naphthalen-1-yl-1,3-oxazole-4-carbonitrile; 5-(propylamino)-2-naphthalen-1-y1-1,3-oxazole-4-carbonitrile; or 5-(butylamino)-2-naphthalen-1-y1-1,3-oxazole-4-carbonitrile.
11. Use of a compound of any one of claims 8-10 for inhibiting 12/15-lipoxygenase.
CA2924141A 2013-08-22 2014-08-22 5-amino 4-cyano substituted oxazole and thiazole derivatives as inhibitors of human 12/15-lipoxygenase Active CA2924141C (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201361868611P 2013-08-22 2013-08-22
US61/868,611 2013-08-22
PCT/US2014/052269 WO2015027146A1 (en) 2013-08-22 2014-08-22 Inhibitors of human 12/15-lipoxygenase

Publications (2)

Publication Number Publication Date
CA2924141A1 CA2924141A1 (en) 2015-02-26
CA2924141C true CA2924141C (en) 2022-06-07

Family

ID=52484189

Family Applications (1)

Application Number Title Priority Date Filing Date
CA2924141A Active CA2924141C (en) 2013-08-22 2014-08-22 5-amino 4-cyano substituted oxazole and thiazole derivatives as inhibitors of human 12/15-lipoxygenase

Country Status (4)

Country Link
US (1) US10287279B2 (en)
EP (1) EP3036226B1 (en)
CA (1) CA2924141C (en)
WO (1) WO2015027146A1 (en)

Families Citing this family (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019060298A1 (en) 2017-09-19 2019-03-28 Neuroenhancement Lab, LLC Method and apparatus for neuroenhancement
US11717686B2 (en) 2017-12-04 2023-08-08 Neuroenhancement Lab, LLC Method and apparatus for neuroenhancement to facilitate learning and performance
EP3731749A4 (en) 2017-12-31 2022-07-27 Neuroenhancement Lab, LLC System and method for neuroenhancement to enhance emotional response
US11364361B2 (en) 2018-04-20 2022-06-21 Neuroenhancement Lab, LLC System and method for inducing sleep by transplanting mental states
CA3112564A1 (en) 2018-09-14 2020-03-19 Neuroenhancement Lab, LLC System and method of improving sleep
US11116778B2 (en) 2019-01-15 2021-09-14 Empirico Inc. Prodrugs of ALOX-15 inhibitors and methods of using the same
US11786694B2 (en) 2019-05-24 2023-10-17 NeuroLight, Inc. Device, method, and app for facilitating sleep
EP4333837A1 (en) * 2021-05-07 2024-03-13 The Regents of the University of California Compounds for modulating platelet-type 12-(s)-lipoxygenase and methods of use for same
CA3228721A1 (en) * 2021-08-09 2023-02-16 The General Hospital Corporation Inhibitors of 12/15-lipoxygenase
WO2024019959A1 (en) * 2022-07-18 2024-01-25 The Regents Of The University Of California Compositions for modulating lipoxygenase and methods of using same

Family Cites Families (84)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US353270A (en) 1886-11-23 Rolling-mill
US3536809A (en) 1969-02-17 1970-10-27 Alza Corp Medication method
US3598123A (en) 1969-04-01 1971-08-10 Alza Corp Bandage for administering drugs
US3773919A (en) 1969-10-23 1973-11-20 Du Pont Polylactide-drug mixtures
US3845770A (en) 1972-06-05 1974-11-05 Alza Corp Osmatic dispensing device for releasing beneficial agent
US3916899A (en) 1973-04-25 1975-11-04 Alza Corp Osmotic dispensing device with maximum and minimum sizes for the passageway
US4008719A (en) 1976-02-02 1977-02-22 Alza Corporation Osmotic system having laminar arrangement for programming delivery of active agent
US4747825A (en) 1984-06-29 1988-05-31 Ferring Laboratories, Inc. Apparatus and methodology for pulsed administration of growth promoting agents
US4834978A (en) 1984-10-01 1989-05-30 Biotek, Inc. Method of transdermal drug delivery
US4687481A (en) 1984-10-01 1987-08-18 Biotek, Inc. Transdermal drug delivery system
US4624665A (en) 1984-10-01 1986-11-25 Biotek, Inc. Method of transdermal drug delivery
US4927687A (en) 1984-10-01 1990-05-22 Biotek, Inc. Sustained release transdermal drug delivery composition
US4810499A (en) 1984-10-01 1989-03-07 Biotek, Inc. Transdermal drug delivery system and method
US4655767A (en) 1984-10-29 1987-04-07 Dow Corning Corporation Transdermal drug delivery devices with amine-resistant silicone adhesives
IE58110B1 (en) 1984-10-30 1993-07-14 Elan Corp Plc Controlled release powder and process for its preparation
US4880633A (en) 1986-03-12 1989-11-14 Merck & Co., Inc. Transdermal drug delivery system
US4797284A (en) 1986-03-12 1989-01-10 Merck & Co., Inc. Transdermal drug delivery system
US4948592A (en) 1986-05-09 1990-08-14 Alza Corporation Pulsed drug delivery
US4723958A (en) 1986-05-23 1988-02-09 Merck & Co., Inc. Pulsatile drug delivery system
US5560922A (en) 1986-05-30 1996-10-01 Rutgers, The State University Of New Jersey Transdermal absorption dosage unit using a polyacrylate adhesive polymer and process
US5163899A (en) 1987-03-20 1992-11-17 Drug Delivery Systems Inc. Transdermal drug delivery system
US4965251A (en) 1987-04-03 1990-10-23 The Board Of Regents Of The University Of Washington Pulse treatment of hemoglobinopathies with erythropoietin
US5312325A (en) 1987-05-28 1994-05-17 Drug Delivery Systems Inc Pulsating transdermal drug delivery system
US5035894A (en) 1987-10-15 1991-07-30 Dow Corning Corporation Controlled release compositions and transdermal drug delivery device
US4917895A (en) 1987-11-02 1990-04-17 Alza Corporation Transdermal drug delivery device
US5405486A (en) 1988-03-04 1995-04-11 Noven Pharmaceuticals, Inc. Apparatus for forming a transdermal drug device
US4877618A (en) 1988-03-18 1989-10-31 Reed Jr Fred D Transdermal drug delivery device
US5073543A (en) 1988-07-21 1991-12-17 G. D. Searle & Co. Controlled release formulations of trophic factors in ganglioside-lipsome vehicle
IT1229203B (en) 1989-03-22 1991-07-25 Bioresearch Spa USE OF 5 METHYLTHETRAHYDROPHOLIC ACID, 5 FORMYLTHETRAHYDROPHOLIC ACID AND THEIR PHARMACEUTICALLY ACCEPTABLE SALTS FOR THE PREPARATION OF PHARMACEUTICAL COMPOSITIONS IN THE FORM OF CONTROLLED RELEASE ACTIVE IN THE THERAPY OF MENTAL AND ORGANIC DISORDERS.
US4956171A (en) 1989-07-21 1990-09-11 Paco Pharmaceutical Services, Inc. Transdermal drug delivery using a dual permeation enhancer and method of performing the same
US5091186A (en) 1989-08-15 1992-02-25 Cygnus Therapeutic Systems Biphasic transdermal drug delivery device
US5252334A (en) 1989-09-08 1993-10-12 Cygnus Therapeutic Systems Solid matrix system for transdermal drug delivery
US5120548A (en) 1989-11-07 1992-06-09 Merck & Co., Inc. Swelling modulated polymeric drug delivery device
US5733566A (en) 1990-05-15 1998-03-31 Alkermes Controlled Therapeutics Inc. Ii Controlled release of antiparasitic agents in animals
US5232702A (en) 1991-07-22 1993-08-03 Dow Corning Corporation Silicone pressure sensitive adhesive compositons for transdermal drug delivery devices and related medical devices
US5234690A (en) 1991-08-23 1993-08-10 Cygnus Therapeutic Systems Transdermal drug delivery device using an unfilled microporous membrane to achieve delayed onset
US5273755A (en) 1991-08-23 1993-12-28 Cygnus Therapeutic Systems Transdermal drug delivery device using a polymer-filled microporous membrane to achieve delayed onset
US5273756A (en) 1991-08-23 1993-12-28 Cygnus Therapeutic Systems Transdermal drug delivery device using a membrane-protected microporous membrane to achieve delayed onset
US5356632A (en) 1991-09-12 1994-10-18 S.I. Scientific Innovations Ltd. Transdermal drug delivery device
US5580578A (en) 1992-01-27 1996-12-03 Euro-Celtique, S.A. Controlled release formulations coated with aqueous dispersions of acrylic polymers
US5308625A (en) 1992-09-02 1994-05-03 Cygnus Therapeutic Systems Enhancement of transdermal drug delivery using monoalkyl phosphates and other absorption promoters
US5358715A (en) 1992-09-02 1994-10-25 Cygnus Therapeutic Systems Enhancement of transdermal drug delivery using monoalkyl phosphates and other absorption promoters
US5421816A (en) 1992-10-14 1995-06-06 Endodermic Medical Technologies Company Ultrasonic transdermal drug delivery system
US5403590A (en) 1992-12-21 1995-04-04 New England Deaconess Hospital Corporation Method of pulsatile drug infusion
US5591767A (en) 1993-01-25 1997-01-07 Pharmetrix Corporation Liquid reservoir transdermal patch for the administration of ketorolac
US5554381A (en) 1993-08-09 1996-09-10 Cygnus, Inc. Low flux matrix system for delivering potent drugs transdermally
US5814599A (en) 1995-08-04 1998-09-29 Massachusetts Insitiute Of Technology Transdermal delivery of encapsulated drugs
US5494680A (en) 1993-12-08 1996-02-27 Minnesota Mining And Manufacturing Company Transdermal delivery device
US5466465A (en) 1993-12-30 1995-11-14 Harrogate Holdings, Limited Transdermal drug delivery system
IT1270594B (en) 1994-07-07 1997-05-07 Recordati Chem Pharm CONTROLLED RELEASE PHARMACEUTICAL COMPOSITION OF LIQUID SUSPENSION MOGUISTEIN
GB2290964A (en) 1994-07-08 1996-01-17 Arto Olavi Urtti Transdermal drug delivery system
US5505958A (en) 1994-10-31 1996-04-09 Algos Pharmaceutical Corporation Transdermal drug delivery device and method for its manufacture
US5879322A (en) 1995-03-24 1999-03-09 Alza Corporation Self-contained transdermal drug delivery device
KR0182543B1 (en) 1995-04-07 1999-03-20 김광호 Illuminating system of microwave oven
US5741511A (en) 1995-04-12 1998-04-21 Sam Yang Co., Ltd. Transdermal drug delivery device for treating erectile dysfunction
US5698217A (en) 1995-05-31 1997-12-16 Minnesota Mining And Manufacturing Company Transdermal drug delivery device containing a desiccant
US5780058A (en) 1995-07-21 1998-07-14 Alza Corporation Oral delivery of discrete units
US5906830A (en) 1995-09-08 1999-05-25 Cygnus, Inc. Supersaturated transdermal drug delivery systems, and methods for manufacturing the same
US5833647A (en) 1995-10-10 1998-11-10 The Penn State Research Foundation Hydrogels or lipogels with enhanced mass transfer for transdermal drug delivery
US6132420A (en) 1996-02-02 2000-10-17 Alza Corporation Osmotic delivery system and method for enhancing start-up and performance of osmotic delivery systems
MY125849A (en) 1997-07-25 2006-08-30 Alza Corp Osmotic delivery system, osmotic delivery system semipermeable body assembly, and method for controlling delivery rate of beneficial agents from osmotic delivery systems
AU1623099A (en) 1997-12-22 1999-07-12 Alza Corporation Rate controlling membranes for controlled drug delivery devices
JP4173635B2 (en) 1997-12-29 2008-10-29 インターシア セラピューティクス,インコーポレイティド Permeation supply apparatus having membrane plug holding mechanism
ATE322928T1 (en) 1997-12-31 2006-04-15 Alza Corp METHOD FOR MONITORING AN OSMOTIC DRUG DELIVERY DEVICE
US6245357B1 (en) 1998-03-06 2001-06-12 Alza Corporation Extended release dosage form
US6365185B1 (en) 1998-03-26 2002-04-02 University Of Cincinnati Self-destructing, controlled release peroral drug delivery system
NZ511465A (en) 1998-11-02 2003-10-31 Alza Corp Controlled delivery of active agents
US6342249B1 (en) 1998-12-23 2002-01-29 Alza Corporation Controlled release liquid active agent formulation dosage forms
CN100410388C (en) 2002-02-08 2008-08-13 霍夫曼-拉罗奇有限公司 Therapeutic and preventing methods for bone loss
US7432271B2 (en) 2003-09-02 2008-10-07 Bristol-Myers Squibb Company Pyrazolyl inhibitors of 15-lipoxygenase
US7351727B2 (en) 2003-09-02 2008-04-01 Bristol-Myers Squibb Company Inhibitors of 15-lipoxygenase
WO2006044556A2 (en) 2004-10-14 2006-04-27 Galileo Pharmaceuticals, Inc. Dual inhibitors of lipoxygenase for treating diabetes
WO2006065686A2 (en) 2004-12-13 2006-06-22 Galileo Pharmaceuticals, Inc. Spiro derivatives as lipoxygenase inhibitors
EP1856040A4 (en) 2005-02-25 2009-09-23 Lilly Co Eli Novel lipoxygenase inhibitors
EP1856079A4 (en) 2005-02-25 2008-12-31 Galileo Pharmaceuticals Inc Spiro-heterocyclic chromans, thiochromans and dihydroquinolines
FR2896694A1 (en) 2006-01-30 2007-08-03 Genfit S A USE OF 15-LIPOXYGENASE INHIBITORS IN THE TREATMENT OF METABOLIC SYNDROME
WO2009017825A2 (en) 2007-08-02 2009-02-05 The General Hospital Corporation Novel lipoxygenase inhibitors as neuroprotective agents
CA2720516C (en) * 2008-04-29 2013-10-01 Joyant Pharmaceuticals, Inc. Indoline anti-cancer agents
US20100248365A1 (en) * 2009-03-27 2010-09-30 Zacharon Pharmaceuticals, Inc. Ganglioside biosynthesis modulators
WO2010118347A2 (en) * 2009-04-10 2010-10-14 Zacharon Pharmaceuticals, Inc. O-linked glycan biosynthesis modulators
WO2011028651A1 (en) * 2009-09-01 2011-03-10 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Inhibitors of human 15-lipoxygenase-1
US20130131140A1 (en) 2010-01-14 2013-05-23 Jerry L. Nadler Treatment of diabetes and disorders associated with visceral obesity with inhibitors of human arachidonate 12 lipoxygenase and arachidonate 15-lipoxygenase
WO2012075393A2 (en) * 2010-12-02 2012-06-07 President And Fellows Of Harvard College Activators of proteasomal degradation and uses thereof
PT2714668T (en) * 2011-05-23 2017-04-26 Merck Patent Gmbh Thiazole derivatives

Also Published As

Publication number Publication date
US10287279B2 (en) 2019-05-14
US20160168137A1 (en) 2016-06-16
EP3036226A1 (en) 2016-06-29
EP3036226A4 (en) 2017-04-05
WO2015027146A1 (en) 2015-02-26
EP3036226B1 (en) 2020-01-08
CA2924141A1 (en) 2015-02-26

Similar Documents

Publication Publication Date Title
CA2924141C (en) 5-amino 4-cyano substituted oxazole and thiazole derivatives as inhibitors of human 12/15-lipoxygenase
JP4344607B2 (en) Pyrrolopyrimidines as protein kinase inhibitors
Liu et al. Design, synthesis and biological evaluation of harmine derivatives as potent GSK-3β/DYRK1A dual inhibitors for the treatment of Alzheimer's disease
JP6786566B2 (en) Heteroaryl compounds and how to use them
Jismy et al. Pyrimido [1, 2-b] indazole derivatives: Selective inhibitors of human monoamine oxidase B with neuroprotective activity
Duan et al. Design, synthesis, and biological evaluation of novel dual inhibitors targeting lysine specific demethylase 1 (LSD1) and histone deacetylases (HDAC) for treatment of gastric cancer
CA3022395A1 (en) Di-substituted pyrazole compounds for the treatment of diseases
AU2017368286B2 (en) 3-amino-1,5-dihydro-pyrazolo[3,4-d]pyrimidin-4-ones as cyclin dependent kinase inhibitors
Li et al. Development of selective HDAC6 inhibitors with in vitro and in vivo anti-multiple myeloma activity
Gao et al. Identification and preliminary structure–activity relationships of 1-Indanone derivatives as novel indoleamine-2, 3-dioxygenase 1 (IDO1) inhibitors
Xiong et al. Development of potent NEDD8-activating enzyme inhibitors bearing a pyrimidotriazole scaffold
TW201643143A (en) Compound for inhibiting neuronal cell death induced by oxidative stress
US20200024285A1 (en) Novel chromone oxime derivative and its use as allosteric modulator of metabotropic glutamate receptors
Ayoup et al. Repurposing 1, 2, 4-oxadiazoles as SARS-CoV-2 PLpro inhibitors and investigation of their possible viral entry blockade potential
Wang et al. Enhancing monoamine oxidase B inhibitory activity via chiral fluorination: Structure-activity relationship, biological evaluation, and molecular docking study
Stylianaki et al. “Hit” to lead optimization and chemoinformatic studies for a new series of Autotaxin inhibitors
US11472806B2 (en) Substituted heterocyclic compounds as allosteric modulators of group II metabotropic glutamate receptors
US11427590B2 (en) Small molecule inhibitors of neutral sphingomyelinase 2 (nSMase2) for the treatment of neurodegenerative diseases
WO2018084266A1 (en) Furanone derivates and methods of use thereof
Codony Gisbert et al. Discovery and In Vivo Proof of Concept of a Highly Potent Dual Inhibitor of Soluble Epoxide Hydrolase and Acetylcholinesterase for the Treatment of Alzheimer's Disease
Champiré et al. Tetrahydropyridine LIMK inhibitors: Structure activity studies and biological characterization
WO2023146880A1 (en) Functionalized allopurinol derivatives for treatment of alzheimer&#39;s disease
WO2023042177A1 (en) Enantiomers of 5-((7-chloroisoquinolin-1-yl)amino)-n-(6-methoxy-1,2,3,4-tetrahydronaphthalen-2-yl)picolinamide
Valencia Exploring the N-Benzylpiperidine and N. N-Dibenzyl (N-Methyl) amine fragments as privileged structures in the search of new multitarget directed drugs for Alzheimer's disease
EP3381897A1 (en) Derivatives of the disodium 2,2&#39;-{carbonylbis[imino-3,1-phenylenecarbonylimino(1-methyl-1h-pyrrole-4,2-diyl)carbonylimino]}dinaphthalene-1,5-disulfonate salt and related compounds as heparanase inhibitors for the treatment of cancer

Legal Events

Date Code Title Description
EEER Examination request

Effective date: 20190626